Received: 18 December 2020 | Accepted: 27 December 2020 DOI: 10.1111/imr.12951

INVITED REVIEW

Host- directed therapy to combat mycobacterial infections*

Gül Kilinç | Anno Saris | Tom H. M. Ottenhoff | Mariëlle C. Haks

Department of Infectious Diseases, Leiden University Medical Center, Leiden, The Abstract Netherlands Upon infection, mycobacteria, such as Mycobacterium tuberculosis (Mtb) and nontu-

Correspondence berculous mycobacteria (NTM), are recognized by host innate immune cells, triggering Mariëlle C. Haks, Department of a series of intracellular processes that promote mycobacterial killing. Mycobacteria, Infectious Diseases, Leiden University Medical Center, Albinusdreef 2, ZA Leiden however, have developed multiple counter- strategies to persist and survive inside host 2333, The Netherlands. cells. By manipulating host effector mechanisms, including phagosome maturation, Email: [email protected] vacuolar escape, , antigen presentation, and metabolic pathways, patho- Funding information genic mycobacteria are able to establish long-lasting infection. Counteracting these Netherlands Organisation for Scientific Research (NWO- TTW), Grant/Award mycobacteria- induced host modifying mechanisms can be accomplished by host- Number: 16444; European Union (Horizon directed therapeutic (HDT) strategies. HDTs offer several major advantages compared 2020 programme), Grant/Award Number: 847762; IMI2 JU (AMR Accelerator to conventional antibiotics: (a) HDTs can be effective against both drug- resistant and program), Grant/Award Number: 853932 drug- susceptible bacteria, as well as potentially dormant mycobacteria; (b) HDTs are and 853903 less likely to induce bacterial drug resistance; and (c) HDTs could synergize with, or shorten antibiotic treatment by targeting different pathways. In this review, we will explore host- pathogen interactions that have been identified for Mtb for which poten- tial HDTs impacting both innate and adaptive immunity are available, and outline those worthy of future research. We will also discuss possibilities to target NTM infection by HDT, although current knowledge regarding host- pathogen interactions for NTM is limited compared to Mtb. Finally, we speculate that combinatorial HDT strategies can potentially synergize to achieve optimal mycobacterial host immune control.

KEYWORDS drug resistance, host- directed therapy, Mycobacterium avium, Mycobacterium tuberculosis, nontuberculous mycobacteria

1 | INTRODUCTION of deaths globally.1 Approximately a quarter of the world's popula- tion is infected with Mtb and in most cases, progression toward TB Tuberculosis (TB), caused by Mycobacterium tuberculosis (Mtb), re- disease is prevented by an efficient host immune response, often mains a major health problem. With an estimated 10 million disease resulting in a latent TB infection (LTBI).1 Five to fifteen percent of cases and 1.4 million deaths in 2019, Mtb is the deadliest infec- LTBI individuals will develop TB disease during their life- time, often tious agent worldwide and TB is one of the top- 10 leading causes concomitant with host immunocompromising conditions, including

This article is part of a series of reviews covering Immunity to Mycobacteria appearing in HIV infection and use of immunosuppressive medication. Treatment Volume 301 of Immunological Reviews. of patients with active TB has largely remained unchanged for over Kilinç, Saris, Ottenhoff, and Haks authors are Contributed equally. 30 years,1 and due to its lengthiness (6- 24 months) and considerable

This is an open access article under the terms of the Creative Commons Attribution-NonCommercial-NoDerivs License, which permits use and distribution in any medium, provided the original work is properly cited, the use is non-commercial and no modifications or adaptations are made. © 2021 The Authors. Immunological Reviews published by John Wiley & Sons Ltd.

Immunological Reviews. 2021;00:1–22. wileyonlinelibrary.com/journal/imr | 1 2 | KILINÇ et aL side effects, treatment- adherence is low fueling development of regimens, thereby increasing compliance. Fifth, HDT may permit multi- drug and extensive- drug resistance (MDR and XDR). The large dose lowering of standard antibiotics, thus reducing toxicity with- TB disease burden and the increasing incidence of drug resistance out impacting efficacy. Finally, as HDT and mycobacterium- targeting make alternative treatment solutions imperative. compounds (ie, antibiotics) by definition act on different pathways, While the number of TB cases is slowly declining, a trend that combinatorial regimens would be expected to synergize. In this re- may well be broken as a result of the COVID- 19 pandemic,2 the prev- view, we will provide a comprehensive overview of host- pathogen alence of infections known to be caused by nontuberculous myco- interactions that have been identified in Mtb infections and that are bacteria (NTM) is increasing at an alarming rate, currently reaching amenable to targeting by HDTs (summarized in Figure 1 and Table 1). 0.2- 9.8 per 100.000 individuals.3 NTM represent a group of op- Furthermore, despite a limited number of reports, we will also dis- portunistic mycobacterial pathogens that mostly cause pulmonary cuss NTM- mediated host modulation and speculate whether HDTs diseases (PD), predominantly in vulnerable populations due to immu- could also be of interest to combat these mycobacterial infections. nodeficiencies and/or pre- existing lung conditions. Mycobacterium Finally, we will discuss the possibility of combinatorial HDTs that tar- avium (Mav) complex (MAC) and Mycobacterium abscessus (Mab) ac- get distinct host signaling pathways to promote possible synergistic count for the large majority of reported cases.3 Despite extended treatment effects. treatment regimens, clinical outcome is poor, with cure rates of approximately 50%- 88% among MAC- PD patients and 25%- 58% among Mab- infected individuals,3 urging the development of novel 2 | HDT MODULATING INNATE IMMUNE treatment modalities. CELL FUNCTION Mycobacteria are well known for their capability to manipulate intracellular signaling pathways to escape from host- defense mech- 2.1 | Phagocytosis and phagosome maturation anisms in human cells. Mtb is best studied in this regard, but NTM have also been shown to modulate host immune responses, includ- The first potential target for HDT to interfere with host- pathogen in- ing preventing phagosome acidification and maturation or escap- teractions is to modulate mycobacterial host- cell entry. Mycobacteria ing from phagosomes into the nutrient- rich cytosol. Counteracting infect host cells, predominantly alveolar and epithe- pathogen- induced immune modulation by host- directed therapy lial cells, in the lower respiratory tract, following inhalation of small (HDT) is a promising adjunct therapy to antibiotic therapy to combat bacteria- containing aerosols. Mycobacteria express pathogen- intracellular mycobacterial infections, with several major advantages associated molecular patterns (PAMPs) that are recognized by pat- over current antibiotics. First, HDT can also be effective against tern recognition receptors (PRRs), such as Toll- like receptors (TLRs), MDR/XDR mycobacteria that are insensitive to current standard C- type lectin receptors, and other scavenger receptors expressed on antibiotics. Second, because there is no direct selection pressure on the surface of host cells to initiate phagocytosis.4 Especially TLR2, mycobacteria, host- targeting compounds are less likely to result in which forms heterodimers with TLR1 and TLR6 to recognize lipo- drug resistance. Third, host- targeting compounds have the potential or lipopeptides (eg, lipomannan), and TLR4, which recog- to target metabolically inactive, non- replicating bacilli during LTBI, nizes cell wall lipids, glycoproteins, and secreted proteins, are known which are tolerant or resistant to conventional therapies. Fourth, to mediate Mtb- induced cellular activation.5 Mice lacking TLR2 are HDT may allow shortening of current lengthy TB/NTM- treatment more susceptible to infections with virulent Mtb and Mav strains.6- 8

FIGURE 1 Host- pathogen interactions and potential host- directed therapies (HDT). Granulomas are characteristic for tuberculosis and mycobacterial infections in general. Pathologic granulomas are poorly vascularized due to ineffective , leading to hypoxia and concomitant host- cell necrosis and bacterial dissemination. Blocking angiogenesis, preventing host- cell necrosis (or stimulating ) or inhibiting extracellular matrix (ECM) degradation improves granuloma structure and concomitant disease outcome. Macrophages, key cells in the anti- mycobacterial response, initiate phagocytosis after toll- like receptor (TLR) recognition, which is prevented and/or modulated by mycobacteria. Promoting TLR4 engagement, TLR2 signaling and post- phagocytic signaling via receptor tyrosine kinase are all potential targets for HDT to improve host immunity during mycobacterial infection. After internalization, mycobacteria are located to phagosomes that slowly mature and ultimately fuse with lysosomes, which are all inhibited by mycobacteria. Alternatively, mycobacteria escape to the cytosol where they can be recognized by cytoplasmic pathogen recognition receptor (PRR) and “recaptured” using autophagy, which again is inhibited by mycobacteria. HDTs that (1) prevent phagosomal escape, (2) alleviate blockage of (auto- )phagosome maturation, (3) promote autophagy and/or (4) stimulate (auto- )phagolysosome fusion all enhance mycobacterial killing. HDT that enhance cytoplasmic recognition of mycobacteria also improve the anti- mycobacterial immune response. Mycobacteria that remain in the cytosol impair host metabolic pathways by stimulating tricarboxylic acid (TCA) cycle intermediates from mitochondria to be expelled into the cytosol to form lipid droplets and induce mitochondrial membrane depolarization. HDTs that (1) impair lipid droplet accumulation, (2) prevent mitochondrial membrane depolarization, and/or (3) stimulate TCA cycle intermediates being allocated in eicosanoid signaling maintain functionality which leads to better mycobacterial control. Finally, mycobacteria prevent the host from mounting an effective adaptive immune response by inhibiting antigen presentation and impairing T- cell skewing. HDTs that promote adaptive immunity by enhancing antigen presentation, stimulating Th1 skewing or inhibiting Th2/Treg immunity all improve disease outcome. Compounds that can correct the above processes are represented in red for inhibitory/blocking therapies and in green for stimulatory therapies and summarized in Table 1, ordered per physiological process KILINÇ et aL | 3

Inhibit MMP and/or ECM degradation

Stimulate Th1 immunity Block angiogenesis (and mediated signalling)

Granuloma

Angiogenesis

Host-cell death

Prevent host-cell necrosis and ferroptosis or stimulate apoptosis

Phagocytosis Promote TLR2 and Promote TLR4 engagement post-phagocytic signaling

Enhance cytoplasmic pathogen recognition

Stimulate host-protective epigentics N N

F L

R

- κ

P B

3 Intracytoplasmic pathogen Restore immune balance by: Stimulate phagosome maturation recognition Eicanosoid limiting immunity via IL-1/prostaglandin signaling signaling Phagosome IL-1 or promoting immunity via type I IFN/leukotriene signaling maturation family

Phagosomal Leukotrienes Prostaglandins escape Type I IFN TCA Promote autophagy intermediates Autophagy/ Inhibit Th2 immunity Xenophagy Metabolism Inhibit lipid droplet accumulation Stimulate Th1 immunity (Auto-)phago Phagosomal lysosome fusion escape

RIG-I-like / NOD Stimulatory HDT receptors Prevent Co-stimulatory Inhibitory HDT phagosomal escape Mycobacteria- Co-stimulatory mediated inhibition receptor Prevent mitochondrial Macrophage dysfunction TLR4 Antigen TLR2 Th1/Th1*/Th17 T cell T cell activation presentation TCR and skewing

MHC class II/I proton pump Dendritic cell IFN-y IL-17 Neutrophil IL-23 Lysosome IL-1β Lipid droplet IL-4 Enhance antigen TCA cycle presentation IL-24 Mitochondria with IL-12 functional OXPHOS Stimulate (auto-)phago- Depolarized lysosome fusion ECM mitochondria

NTM, including Mav and Mab, express a class of glycolipids known TLR2- MyD88 pathway has been linked to phagolysosome escape of as glycopeptidolipids (GPLs) which mask underlying cell wall virulent Mtb to the cytosol,11,12 indicating a crucial role for TLR sign- phosphatidyl- myo- inositol mannosides, thereby limiting interactions aling pathways in the control of intracellular mycobacteria. Several with TLR2.9,10 Moreover, the acetylation state of lipomannan modu- PRR agonists, including TLR2 agonist Pam2Cys, have been identified lates TLR2- mediated macrophage activation, and subversion of the that activate both innate and adaptive immune responses against 4 | KILINÇ et aL 140 (Continues) 160 149 160,188,191 121 116 122 13 124 129 133,134,137- 141 142 145 131 146- 151 152 158- 155 99 161 161 157- Ref. Mtb Mtb Mtb Mtb Mtb Mtb Mtb Mtb, Mav Mtb Mtb Mtb Mtb Mtb, Mav Mtb Mtb Mtb Mtb Mtb Mtb Pathogen Mtb in vitro ex vivo, in vivo ex vivo, in vivo in vitro ex vivo, in vivo in vitro, in vivo in vitro, in vivo in vitro, (R)CT in vivo in vivo in vivo in vitro in vivo, (R)CT in vivo CR (R)CT in vitro in vitro in vitro, in vivo, R(CT) Model in vitro α α signaling) signaling) ↑ Th1 immunity↑ Th1 ↑ Th1 immunity↑ Th1 ↑ Antigen presentation ↑ Antigen presentation immunity↑ Th1 (TLR4 immunity↑ Th1 (TLR4 immunity↑ Th1 immunity↑ Th1 immunity↑ Th1 ↓ Immune response ↓ Th2 immunity immunity↑ Th1 immunity↑ Th1 immunity↑ Th1 ↓ Immune response ↑ Apoptosis ↓ Necrosis ↑ TNF- ↓ Necrosis ↓ Immune response Biological activity ↑ TNF- Cells 4 cytokines

+ 106binhib. a

CD40 agonist HMBPP

ZM1 γ 1 inhib. + +

4A

89 1MT 24 2 2 12 Bergenin MiRNA- AGK2 G1- LPS D- Skewing of T- IFN- IL- IL- FPS- Blocking IL- IL- IL- PBMCs Prednisolone (PROGRAMMED) cell death Mcl- Nicotinamide SQ22536 Dexamethasone Compound Antigen presentation and priming H- 186 224 47,51,52 92 17 77 13- 19 20 26,27 28 29 29 29 33,34 71- 78 84,222- 85 86- 70 70 187 181,184- 26,27 40- Ref. Mtb Mtb Mtb Mtb, Mm Mtb Mtb Mtb Mtb Mtb, Mav Mtb Mtb Mtb Mtb Pathogen Mtb Mtb Mtb Mtb Mtb Mtb Mtb (R)CT (R)CT (R)CT CR (R)CT in vitro, in vivo, in vitro, in vivo, in vitro in vitro, in vivo, in vitro in vivo, (R)CT in vitro in vitro, in vivo in vitro, in vivo, in vitro, in vivo in vitro in vitro in vitro, in vivo Model in vitro in vivo in vitro in vitro in vitro, (R)CT in vitro, (R)CT in vitro, in vivo, phagolysosome phagolysosome phagolysosome phagolysosome phagolysosome fusion fusion fusion fusion fusion recognition ↑ Phagosome maturation ↑ (Auto)- ↑ TLR2 signaling ↑ Phagosomal acidification ↑ Autophagy ↑ Phagocytosis ↓ Phagocytosis Unknown ↑ (Auto)- ↑ Phagocytosis ↑ (Auto)- ↑ Lysosomal biogenesis ↑ (Auto)- ↑ TLR2 signaling ↑ (auto)- Biological activity ↑ PRR signaling ↑ Phagosomal acidification Unknown Unknown ↑ Cytoplasmic pathogen ↑ Autophagy ↑ Phagosome maturation

a

a

5p inhib.

a

a a

HDT compounds HDT and their biological activity against mycobacterial infections

a

a

a a D a a 23a- 125 inhib.

2076 a

a CSF ENMD- SRT1720 Compound Phagocytosis and phagosome maturation PRR agonists miRNA- Imatinib AZD0530 AT9283 Dovitinib Nitazoxanide Metformin Gefitinib Bedaquiline 2062 GM- Resveratrol Celecoxib Statins miRNA- Autophagy Imatinib Vitamin- TABLE 1 KILINÇ et aL | 5 186 (Continues) 157 163 163 164,165 164,165 171 129 176,177 174 175 176,177 180 179 181,184- 189,190 187 187 192,193 Ref. Mm Mtb Mtb Mtb, Mav Mtb Mtb Mm Mtb Mm Mtb Mtb Mtb Mtb Mtb Mtb Mtb Mtb, Mav Pathogen Mtb Mtb in vivo in vivo in vivo in vitro in vitro, in vivo in vitro in vivo in vitro in vivo in vitro in vitro in vivo in vivo, (R)CT in vivo ex vivo, in vivo in vivo, (R)CT in vitro Model in vitro, in vivo, (R)CT in vivo α α α /prostaglandin /prostaglandin /prostaglandin /prostaglandin β β β β 1 1 1 1 dysfunction accumulation signaling signaling signaling signaling incorporation ↑ Apoptosis ↓ Ferroptosis ↓ Necrosis ↓ TNF- ↓ Necrosis ↓ Necrosis ↓ TNF- ↓ TNF- ↓ Glycolysis ↑ Immune response? ↓ Mitochondrial ↓ Lipid droplet ↑ IL- ↓ IL- ↑ IL- ↑ Apoptosis ↓ IL- ↑ Iron chelation Biological activity ↓ Membranal cholesterol ↓ Glycolysis

a a

a

a a 4 Zileuton 1MT DG Ferrostatin 1 Alisporivir Doramapimod Desipramine Cilostazol Sildenafil D- administrationATP Carbohydrate and lipids 2- administrationATP Clemastine M1 Ezetimibe Statins Eicosanoids PGE2 and/or Celecoxib LTB Ibuprofen Compound Antigen presentation and priming FX11 186 224 109 62 53 101 77 55- 49- 66 68 69 71- 78 79 80 84,222- 70 70 19 20 181,184- 95,97 99- 107- 79 Ref. Mtb Mtb Mtb Mtb Mtb Mtb Mtb Mtb Mtb Mtb Mtb Mtb Mtb Mtb Pathogen Mtb Mtb Mtb Mtb Mtb Mtb (R)CT (R)CT (R)CT (R)CT (R)CT in vivo in vitro, in vivo, in vitro, in vivo, in vitro, in vivo, in vitro, in vivo, in vitro in vitro in vitro in vitro in vitro, in vivo in vitro, in vivo in vitro in vitro, in vivo in vitro Model in vitro, (R)CT in vitro, (R)CT in vitro, ex vivo in vitro, ex vivo (R)CT in vitro, in vivo, dysfunction ↑ Autophagy ↓ ↑ Autophagy, ↑ Autophagy ↑ Autophagy ↑ Autophagy ↑ Reactive oxygen species ↑ Autophagy ↓ Mitochondrial ↑ Reactive oxygen species ↑ Autophagy ↑ Autophagy ↑ Autophagy ↑ Autophagy ↑ Autophagy Biological activity ↑ Autophagy ↑ Autophagy ↑ Autophagy ↑ Autophagy ↑ Reactive nitrogen species ↑ Autophagy

a

a ) 2+

a a

5p inhib.

a

a a (Continued)

cysteine a a A (± Zn 27a antagomir 27a 23a- 125 inhib.

a

a acetyl- arginine Bedaquiline CD157 Vitamin- Compound Phagocytosis and phagosome maturation PBA MiRNA- Everolimus Ibrutinib Metformin Gefitinib Bazedoxifene Loperamide 2062 Resveratrol SRT1720 miRNA- Statins Intracellular killing mechanisms N- L- Bazedoxifene miRNA- TABLE 1 6 | KILINÇ et aL DG, 198 ; (R)CT,; 193- 208 25 212,213 166 210,214 203,205 205 205 217 218 218 219 Ref. Mtb Mtb Mm Mtb Mtb Mtb Mtb Mtb Mtb Mtb Mm Mm Mm Pathogen Mm, Mycobacterium marinum ; in vivo, (R)CT in vivo in vivo in vitro, in vivo in vivo in vitro, in vivo, (R)CT in vitro, in vivo in vivo in vivo in vivo in vivo in vivo in vivo Model stimulating factor; PBA, phenylbutyrate; SAHA, suberoylanilide Mav, Mycobacterium avium ; /prostaglandin β 1 signaling enyl pyrophosphate; PBMCs, peripheral blood mononuclear cells; 2- ↓ IL- ↓ MMP/ECM degradation↓ MMP/ECM degradation↓ MMP/ECM degradation↓ MMP/ECM degradation↓ MMP/ECM degradation↓ MMP/ECM degradation↓ MMP/ECM degradation↓ MMP/ECM degradation↓ MMP/ECM ↓ Angiogenesis ↓ Angiogenesis ↓ Angiogenesis ↓ Angiogenesis Biological activity 2- macrophage colony- but- methyl- 3- 9 inhib. 9 inhib. 9785 3ct CSF, granulocyte- CSF, Aspirin Granuloma: formation, angiogenesis and hypoxia Cipemastat MMP- Sb- AB0046 Doxycycline Marimastat Batimastat MMP- Bevacizumab SU5416 Pazopanib AKB- Compound Antigen presentation and priming Mtb, Mycobacterium tuberculosis hydroxy- 4- 77 71- 187 115 115 115 70 70 117 117 Ref. ; HMBPP, (E)- HMBPP, ; α Mtb Mtb Mtb Mtb Pathogen Mtb, Mm Mtb, Mm Mtb, Mm Mtb Mtb B4; matrix and MMP, metalloprotease. , - leukotriene- (R)CT α 4, in vitro, in vivo, in vivo, (R)CT in vitro in vitro, in vivo Model in vitro, in vivo in vitro, in vivo in vitro, in vivo in vitro in vitro ; IL,; ; TNF- γ protective protective protective protective protective protective protective epigenetics epigenetics epigenetics epigenetics epigenetics epigenetics epigenetics ↑ Reactive oxygen species ↑ Reactive oxygen species ↑ Host- ↑ Host- Biological activity ↑ Host- ↑ Host- ↑ Host- ↑ Host- ↑ Host- , - γ

a a glucose; ATP, adenosine glucose; ATP, triphosphatases; LTB

(Continued)

a a D- deoxy- Compound Phagocytosis and phagosome maturation Metformin Celecoxib Epigenetic regulation TMP195 TMP269 Trichostatin A Resveratrol SRT1720 Valproic acid SAHA Compounds targeting distinct host intracellular pathways are categorized under multiple sections. (randomized) clinical trial; CR, case report; PRR, pathogen recognition receptor; inhib., inhibitor; GM- TABLE 1 hydroxamic acid; IFN- 2- a KILINÇ et aL | 7

Mtb, positioning PPRs as potential HDT targets to combat mycobac- Formation and subsequent acidification of phagolysosomes is terial infections.13- 17 Furthermore, also downstream PRR signaling also inhibited by Mtb- secreted 1- tuberculosinyladenosine might be modulated by HDT. TLR2- dependent expression of miRNA- antacid (1- TbAd).30 Accumulation of 1- TbAd in acidic intracellular 125 hampered autophagy in murine macrophages.18,19 TLR2- MyD88 compartments resulted in swelling and ultimately bursting of phago- signaling in Mtb- infected murine macrophages was also repressed somes, permitting mycobacterial escape into the cytosol. To further by upregulation of miRNA- 23a- 5p, restricting Mtb infection– induced impair phagosome integrity, Mtb permeabilizes phagosomal mem- autophagy and thus increasing intracellular Mtb survival.20 Inhibition branes using the bacterial ESX- 1 secretion system (ie, ESAT- 6),31 of miRNA- 12518,19 or miRNA- 23a20 both reduced Mtb survival, iden- which leads to leakage of phagosomal cargo into the cytosol, allowing tifying miRNA- 125 and miRNA- 23a as potential targets for HDTs. phagosomal escape of mycobacteria. Although the cytosol contains After phagocytosis, mycobacteria become localized in phago- an abundance of nutrients to support bacterial growth, translocation somes that are initially non- degradative, but slowly mature into into the cytosol also activates DNA- and RNA- sensing pathways via increasingly hostile organelles. This so- called phagosome matura- intracellular recognition of PAMPs and danger- associated molecular tion hinges upon fusion with lysosomes that contain antimicrobial patterns (DAMPs) to induce anti- mycobacterial host effector mech- peptides and induce intravesicular acidification enhancing lysosomal anisms. Retinoid acid– inducible I (RIG- I)– like receptors are cy- enzyme activity.21 While initially thought to be simply transport tosolic PRRs recognizing single- and double- stranded RNA and upon vehicles, phagosomes have appeared to be highly dynamic struc- ligation induce the type- I IFN pathway, among others.32 Nucleotide- tures that are regulated by several membrane markers, such as PI3P, binding oligomerization domain (NOD)– like receptors are intracellu- acidifying proton adenosine triphosphatases (ATPases), and Rab- lar sensors for several DAMPs and PAMPs, including bacterial RNA, GTPases.21,22 As GTPases are also involved in autophagy induction, that can induce both type- I IFN and IL- 1 responses.32 Enhancing ex- these enzymes could be interesting targets for HDT, but as of yet pression levels of RIG- I- like receptors using nitazoxanide treatment have not been investigated in this context. To prevent (auto)phago- during mycobacterial infection increased IFN-β levels and concomi- some maturation, Mtb secretes proteins such as SapM and PknG, tantly reduced mycobacterial loads in an in vitro TB model,33 but did which inhibit PI3P- phosphorylation, dissociation of early endosomal not show efficacy in TB patients, possibly due to negligible concen- protein Rab5 and acquisition of late endosomal protein Rab7.23 In trations at the site of infection.34 addition, Mtb prevents recruitment of the proton pumping enzyme vacuolar- type H+- ATPase (v- ATPase) by phagosomes, thus further arresting phagosome acidification.24 Several receptor tyrosine ki- 2.2 | Autophagy nases (RKTs) that are activated upon internalization of Mtb and NTM are involved in both bacterial uptake and intracellular trafficking, Autophagy is a mechanism mediating self- maintenance and cellular and could be exploited as targets for HDT.25 Abelson tyrosine ki- homeostasis and is induced under stress such as hypoxia, starvation nase (Abl), involved in bacterial uptake by regulating cytoskeletal but also microbial infection.35 Autophagy is crucial during Mtb and dynamics in host cells, can be chemically inhibited by imatinib,24 and NTM infections and inhibition of autophagy using azithromycin in- indeed, imatinib treatment impaired internalization of Mtb by human creased susceptibility of cystic fibrosis patients to NTM infection.36 macrophages.26 Furthermore, Abl also modulates the expression of Autophagy is initiated by formation of a double- membraned v- ATPase, and inhibiting RTKs with imatinib induced expression of phagophore that, under stringent control of ubiquitin- like protein several v- ATPase pump- subunits and their colocalization with Mtb- conjugation systems, expands around the intracytoplasmic cargo containing phagosomes, promoting phagosomal acidification and to form autophagosomes, which ultimately fuse with lysosomes enhancing bacterial killing in human macrophages.27 In line with to mediate degradation. Two autophagic pathways are important this, imatinib treatment of mice infected with Mtb or Mycobacterium for mycobacterial degradation: LC3- associated phagocytosis (LAP) marinum (Mm) decreased mycobacterial load and combining ima- and the STING- dependent cytosolic pathway.37 LAP is initiated by tinib with first- line anti- TB drug rifampicin synergistically reduced downstream signaling of numerous receptors, including TLRs,37 mycobacterial load in mice and in a murine macrophage- like cell after which the phagosome becomes decorated with PI3P produced line.25- 27 The potential of inhibiting host tyrosine kinases to im- by the PI3KC3 complex, that includes Beclin- 1 and Rubicon. PI3P pair intracellular mycobacterial survival is further highlighted with and Rubicon are required for the generation of reactive oxygen AZD0530 treatment, a Src- inhibitor, that lowered disease burden in species (ROS) and conjugation of lipidated LC3- II to the membrane Mtb- infected guinea pigs by promoting phagosomal acidification.28 to enhance phagosomal maturation.37 The STING- dependent path- Moreover, Korbee et al showed that inhibiting RTK signaling with way is triggered by mycobacterial DNA released into the cytosol the repurposed drugs AT9283, ENMD- 2076 and dovitinib signifi- through the bacterial ESX- 1 system. When mycobacterial DNA is cantly reduced intracellular Mtb in primary human macrophages.29 sensed by a STING- dependent DNA sensor, cytosolic Mtb is ubiq- Being repurposed, these compounds are already FDA- approved or uitinated by the ubiquitin- ligating (E3) ligase, bound to autophagic in phase II and III clinical trials, thus accelerating potential clinical receptors including p62/sequestosome 1, NDP52 protein and TBK1, application as adjunct therapy in treating (MDR)- TB and treatment and subsequently delivered to autophagosomes by engagement of of refractory NTM infections. membrane- associated LC3.32 8 | KILINÇ et aL

Numerous drugs have been identified that promote autophagy In addition to Beclin- 1 and TBK1, other components of the au- by targeting different components of the autophagic pathways. tophagic pathways have also been targeted to promote mycobac- Beclin- 1 is induced by human antimicrobial peptide (AMP) LL- 37, terial clearance. Ca2+ signaling is pivotal in inducing autophagy by also known as cathelicidin.38 Cathelicidin is able to suppress Mtb activating the Ca2+/calmodulin- dependent serine/threonine- kinase growth and can be induced by pathogens after TLR2/TLR1 ligation, (CaMKK2)/ULK1 complex.65 CaMKK2- mediated autophagy and kill- and also by vitamin- D.39 In vitro experiments identified calcitriol, the ing of intracellular Mtb requires Ca2+ transporter CACNA2D3 which bioactive metabolite of vitamin- D, to exert antimicrobial activity by is, however, suppressed by Mtb- induced miRNA27a.66 Intracellular mediating intracellular killing of Mtb through cathelicidin.40 Calcitriol survival of Mtb could be impaired by inhibiting miRNA- 27, providing has also been linked to nitric oxide (NO) production and suppression a new HDT target. of matrix metalloproteases (MMPs) which may further protect the Another important negative regulator of autophagy is the PI3K- host from TB immunopathology.41,42 The efficacy of vitamin- D as Akt- mTOR signaling pathway, which is robustly activated by Mtb to HDT during TB disease has been investigated in multiple random- facilitate its intracellular survival.67 Everolimus, an improved analog ized controlled trials (RCTs). Vitamin- D administration corrected of mTOR inhibitor rapamycin, was able to reduce Mtb burden in a any vitamin- D deficiencies and was safe in use but did not show human granuloma model and these effects were additive to first- line consistent beneficial outcomes during mycobacterial infections in TB drugs, possibly by HDT activity and/or by inhibiting mycobacterial meta- analyses.43- 45 Acceleration of Mtb clearance from sputum was growth directly.68 Inhibition of protein- kinase C- beta (PKC- B), another mainly observed in MDR- TB cases or patients with a specific gen- important regulator of the PI3K- Akt- mTOR pathway, by ibrutinib otype, such as polymorphisms in the vitamin- D receptor- gene.46,47 also enhanced autophagy and restricted intracellular growth of Mtb Furthermore, low levels of vitamin- D have been linked to a higher in macrophages and mice in the spleen, although not in the lungs.69 susceptibility to develop TB disease.48 Some studies combined Alleviating the Mtb- mediated suppression of sirtuin- 1, a class- III his- vitamin- D therapy with phenylbutyrate (PBA), which stimulates tone deacetylase that also modulates autophagy via 5’AMP- activated cathelicidin- induced autophagy and also inhibits bacterial growth protein- kinase (AMPK), using resveratrol restricted intracellular Mtb directly.49,50 Combining vitamin-D and PBA treatment further in- growth by stimulating autophagy and phagosome- lysosome fusion.70 creased expression of cathelicidin in healthy volunteers, but the aug- Metformin, a well- established stimulator of AMPK- mediated inhibi- mented expression level was constrained to a defined dose range of tion of mTOR signaling, is widely used for the treatment of type- 2 PBA.51 The narrow therapeutic window of PBA might clarify why diabetes, but also induces ROS production, phagosome maturation, certain RCTs failed to detect accelerated sputum- smear conversion and autophagy in vitro and prevents mitochondrial membrane depo- by co- administering vitamin- D and PBA52 and only showed acceler- larization.71- 73 In non- diabetic healthy volunteers, metformin treat- ated sputum- smear conversion at week 4 following combined treat- ment downregulated involved in Mtb- mediated modulation of ment, but not at week 8 in vitamin- D- deficient patients.53 Due to autophagy, as well as type- I IFN signaling, while upregulating genes these inconsistencies, progression of vitamin- D as potential HDT in involved in phagocytosis and ROS production.74 Several clinical trials TB treatment regimens has not been successful. have shown that metformin treatment reduces the risk of latent TB Vitamin- A- deficiency has also been correlated with an in- reactivation and TB mortality, and in patients with cavitary TB, im- creased risk to develop TB disease.54 STING- dependent autoph- proves sputum culture conversion.75- 77 agy can be targeted by the active metabolite of vitamin- A, that Like metformin, repurposing of drugs that are clinically approved is, all- trans retinoic acid (ATRA), which promotes TBK1- mediated in the context of other diseases has been shown to enhance auto- enhancement of autophagy which reduces Mtb survival in human phagy and to reduce intracellular bacterial growth, suggesting these macrophages.55 ATRA is also known to increase CD1d receptor drugs may be considered as HDT candidates. The anti- drug expression on innate immune cells,56 and treatment with non- gefitinib, an inhibitor of epidermal receptor (EGFR), mycobacterial CD1d ligand α- galactosylceramide (α- GalCer) re- promotes intracellular Mtb killing by alleviating the STAT3- dependent duced mycobacterial load and improved survival of mice with repression of effective immune responses in Mtb- infected mice and 57 TB, and while α- GalCer combined with ATRA and vitamin- D did by enhancing lysosomal biogenesis and targeting of mycobacteria not clear the infection in mice, it improved containment of the in- to lysosomes in Mtb- infected macrophages.78 Gefitinib also induced fection.58 In patients, vitamin- A supplementation combined with autophagy,78 but since no specific targeting of mycobacteria to the Zn2+ or vitamin- D gave inconsistent results.59- 62 Thus, although autophagic pathway was observed, this activity has not been for- vitamin- A reduced Mtb loads in vitro and in vivo, evidence for its mally linked to restricting intracellular Mtb survival. Bazedoxifene, efficacy in patients is inconsistent. An additional regulator of the a selective estrogen receptor modulator (SERM) used for breast selective STING- dependent autophagy pathway is DNA- damage cancer treatment, was also shown to inhibit intracellular Mtb growth regulated autophagy- modulator protein 1 (DRAM1). DRAM1 was in macrophages through enhanced ROS- dependent autophagy79 found to trigger autophagy in both Mtb- infected human mac- and to inhibit Mtb growth in liquid culture. Furthermore, one study rophages and Mm- infected zebrafish larvae, whereas DRAM1- showed that loperamide, an anti- diarrheal drug, promoted autoph- deficiency resulted in host- detrimental cell death, underscoring agy as indicated by p62 degradation and decreased mycobacterial DRAM1 as an interesting target for HDT.63,64 burden in vitro and ex vivo in murine macrophages.80 KILINÇ et aL | 9

Mtb not only inhibits the initiation of autophagy, but also fusion Although ROS production is important for host resistance of autophagosomes with lysosomes via protein P2- PGRS47.23,81 against mycobacterial infection, modulating ROS as HDT strat- Furthermore, the Mtb secretion- factor SapM inhibits Rab7- egy requires careful monitoring as excess ROS leads to oxidative recruitment to prevent autophagosome- lysosome fusion.82 Mtb- stress and concomitant necrosis.98 Corroborating this view, re- expressed mannosylated lipoarabinomannan (ManLAM) also inhibits ducing ROS accumulation in Mtb- infected macrophages with ROS maturation of autophagosomes, by blocking LC3- translocation to scavenger N- acetyl- cysteine in fact restricted Mtb replication and autophagosome membranes.32,83 Releasing such blockades in restored macrophage cell viability,99 and in a guinea pig Mtb infec- autophagosome- lysosome fusion could represent potential HDT tion model N- acetyl- cysteine administration was also shown to be strategies. Bedaquiline, a novel antibiotic now in use for MDR- TB, efficacious.100 Moreover, N- acetyl- cysteine was found to be safe in has also been shown to induce phagosome- lysosome fusion and au- a cohort of TB- HIV co- infected individuals,101 although its impact tophagy via activation of TFEB, possibly contributing to it successful on culture conversion remains to be determined.101 Nevertheless, application as a new TB drug.84 In line with this, a small molecule ROS production is important for the bactericidal activity of macro- called 2062 improved autophagy and lysosomal pathway activity phages102 and the critical balance in ROS production and its regu- via activation of TFEB when administered with suboptimal doses of lation is important in restricting intracellular mycobacterial growth rifampicin.85 without harming the host. Although autophagy- targeting HDTs have been investigated Multiple studies in mice and humans have shown antimicrobial mainly in the context of Mtb infections, several case reports have effects of NO, but the exact underlying mechanisms remain un- been published for (disseminated) Mav infections in patients who clear.103,104 Macrophages from LTBI patients were shown to control received granulocyte- macrophage colony- stimulating factor (GM- Mtb growth via NO production, and human macrophages required 105 CSF) or IFN-γ . GM- CSF treatment during Mtb infection reduced iNOS for intracellular killing of Mtb. Moreover, compared to wild- bacterial burden by promoting phagosome- lysosome fusion and type murine macrophages, protein- kinase R (PKR)– deficient macro- 106 increased expression of TNF-α , IFN-γ , and inducible nitric oxide phages induced higher levels of iNOS during Mtb infection, and synthase (iNOS).86- 88 GM- CSF treatment during Mav infection PKR- deficient mice had lower Mtb loads and less severe lung pathol- increased phagocytosis and impaired bacterial growth in vitro in ogy compared to infected wildtype mice, highlighting the potential human macrophages and in Mav- infected patients with or with- of PKR as HDT target. Despite its importance as substrate for NO out HIV infection (partially) improved clinical outcome.89- 92 Thus, production, supplementing L- arginine did not consistently improve autophagy likely plays an important role also in NTM immunity, clinical outcomes such as cure rate or (sputum) smear conversion in and could represent an attractive target for HDT in severe NTM several clinical trials.107- 109 infections. Several Mtb- associated proteins have been identified that pro- tect Mtb from RNS, but Mav naturally tolerates intracellular NO levels and may even benefit from host NO.110- 112 Mice that cannot 2.3 | Intracellular killing mechanisms: reactive produce NO were more resistant to Mav infections, while being more oxygen and nitrogen species prone to Mtb infections.113 In agreement with this, compared to wildtype mice, NOS2- deficient mice showed higher IFN-γ responses To eliminate mycobacteria during infection, host cells trigger the during Mav infection and increased accumulation of especially CD4+ production of reactive oxygen species (ROS) and reactive nitrogen T- cells.114 Enhancing NO production can thus be beneficial in com- species (RNS), via NADPH oxidase 2 (NOX2)93 and inducible nitric batting mycobacterial infections such as Mtb, but not Mav. oxide synthase 2 (iNOS), respectively. iNOS catalyzes production of nitric oxide (NO) by converting L- arginine into L- citrulline, which is subsequently converted into RNS.94 Once expressed, both ROS 2.4 | Epigenetic regulation and RNS interact with the phagosome to destroy bacterial compo- nents.95 Mycobacterium- induced ROS production occurs via the Macrophage polarization is an important mechanism of the immune TLR(2)- MyD88 signaling axis and impairments in this pathway in- system to respond adequately to the plethora of pathogens, which crease susceptibility to Mtb infection.7,96 Recently, TLR2- dependent is partly mediated by epigenetic regulation of using ROS production and bactericidal activity was found to be impaired histone acetylation. The level of histone acetylation is regulated by in CD157- deficient murine macrophages which could be rescued by the balanced activity of histone acetyltransferases (HATs) and his- administration of soluble CD157.95,97 Moreover, expression levels of tone deacetylases (HDACs). HDACs are divided into four classes, CD157, an enzyme important for leukocyte migration and involved three of which are Zn2+- dependent (class I, IIa/IIb and IV), while in nicotinamide- adenine- dinucleotide (NAD+) metabolism, are ele- class-III is NAD+- dependent.115 Mtb infection actively modulates vated in patients with active TB compared to LTBI and lowered when the acetylation status of host histones by (a) suppressing expres- patients are treated with TB , indicating an important sion of class- II HDACs (ie, HDAC 3, 5, 7, and 10) in macrophages, role of CD157 in host immunity, biomarker profiling and also provid- with anti- inflammatory M2 macrophages being more affected than ing a potential HDT.97 pro- inflammatory M1 macrophages,115 (b) inhibiting the expression 10 | KILINÇ et aL of class- III HDAC sirtuin- 1 both in vitro and in human tissues from MHC-class II. By actively engaging TLR2 rather than other TLRs, Mtb TB patients,70 and (c) upregulating expression of sirtuin- 2, another (and to a lesser extent M smegmatis) minimizes upregulation of CIITA class- III HDAC that regulates cell cycle and metabolism.116 Type IIa– and concomitant MHC- class II expression. In addition, TLR2 (among specific HDAC inhibitors (HDACi) TMP195 or TMP269 reduced bac- all TLRs) most potently induces an innate (IL- 6) response,120 leading terial loads in M2, but not M1 macrophages, while broad- spectrum to upregulation of suppressor- of- - signaling- 1 (SOCS1) that HDACi trichostatin A reduced bacterial loads in both M1 and M2 in turn inhibits signal- transducer- and- activator- of- transcription 1 macrophages. Interestingly, combining HDACi with AKT1 kinase (STAT1) phosphorylation and antigen presentation, further impair- inhibitor H- 89 resulted in cumulative reduction in bacterial loads. ing the adaptive host immune response.23 MiR106b, which degrades Importantly, in a Mm zebrafish infection model, both class- IIa and mRNA encoding cathepsin S, a protein that modulates MHC- class II pan- HDAC inhibition reduced bacterial loads, confirming the in molecules to allow peptide loading, is significantly upregulated dur- vivo potential of HDAC inhibition as HDT to treat TB.115 Sirtuin- 1, ing Mtb infection.121 Inhibition of miR106b using miRIDIAN hairpin a class- III HDAC important during (viral) infections, regulates stress inhibitors upregulated expression of both cathepsin S and HLA-DR responses and cellular metabolism. Resveratrol or SRT1720, a and enhanced subsequent CD4 T- cell proliferation.121 Alternatively, natural and synthetic activator of sirtuin- 1, enhanced clearance of inhibition of sirtuin- 2 activity in macrophages using AGK2 modu- drug- sensitive and drug- resistant Mtb.70 Both compounds stimulate lated gene expression- promoted antigen presentation.116 AGK2 autophagy and phagolysosome fusion in THP- 1 cells, which likely ac- treatment of mice resulted in upregulation of MHC- class II expres- counts for the enhanced bacterial killing, while reducing pathology sion but also of co- stimulatory molecules and markers of activation, 116 in a TB mouse model, possibly by inhibiting expression of IL- 1β, IL- 6, leading to enhanced priming of T- cells and improved Mtb killing. 70 MCP- 1, and TNF-α . Since Mtb limits activation of antigen- presenting cells (APCs), The Mtb genome encodes Rv1151c, a sirtuin- like NAD- which precludes the host from mounting an effective adaptive im- dependent deacetylase, allowing Mtb to produce acetyl coenzyme mune response, proper activation of APCs could be an interesting A (acetyl- CoA) synthetase, a critical enzyme in energy metabo- HDT. A possible strategy for HDT could be administration of G1- lism of both host cells and bacteria. Targeting this pathway using 4A, a polysaccharide from Tinospora cordifolia that presumably sig- HDACi valproic acid directly inhibited bacterial growth, likely by nals via TLR4, or TLR4 ligand LPS combined with a CD40 agonistic inhibiting acetyl- CoA production by Mtb itself, while co- treatment antibody.13,122 Both treatments induced vast cytokine production of valproic acid and rifampicin/isoniazid therapy resulted in cumu- (IFN-γ /IL- 12, TNF-α , IL- 6) and upregulation of co- stimulatory mol- lative effects.117 By contrast, FDA- approved HDACi suberoylanilide ecules by dendritic cells in vitro.122 Furthermore, both treatments hydroxamic acid (SAHA) had no direct effect on Mtb growth, but reduced bacterial loads in murine TB infection models which was, at it reduced mycobacterial growth via host- directed mechanisms and least in part, T- cell- mediated.122 However, systemic administration synergized with rifampicin/isoniazid therapy.117 As Rv1151c is well of TLR ligands is known to cause significant side effects,123 and may conserved across different mycobacterial species including Mav,118 only be applicable via local administration. Bergenin, a phytochemi- the above therapies may also be efficacious against NTM. cal extracted from tender leaves, enhanced macrophage activation, as evidenced by enhanced CD11b expression as well as augmented NO, TNF-α , and IL- 12 production, through activating the MAPK/ 3 | HDT MODULATING ADAPTIVE IMMUNE ERK pathway. The resulting increased IL- 12 production induced a RESPONSES robust Th1 response with concomitant IFN-γ production by T- cells. Bergenin therapy reduced bacterial loads as well as lung pathology 3.1 | Antigen presentation and priming in a murine TB infection model.124 Of note, vaccination could also be an interesting HDT approach to activate APCs or reprogram an Upon phagocytosis, pathogens are processed and degraded, such effective adaptive immune response. This, however, falls outside the that pathogen- derived peptides can be loaded and presented in scope of this review and is excellently reviewed elsewhere.125 MHC- class I and II molecules to initiate adaptive T- cell responses. Due to the chronic immune stimulation during persisting myco- One strategy of mycobacteria to evade host adaptive immune re- bacterial infections, including LTBI, T- cells and APCs upregulate inhib- sponses is to impair presentation of mycobacterial peptides by evad- itory receptors such as PD- 1/PD- L1, which can impair T- cell effector ing phagosomal degradation. Improving mycobacterial degradation functions,23 and may be interesting targets for HDT. Expression by promoting phagosomal maturation and/or autophagy induction of exhaustion- associated markers by T- cells during active disease, as discussed above likely enhances both antigen presentation and however, is rather ambiguous: Despite successes in anti- cancer ther- concomitant adaptive immunity. Another strategy of mycobacte- apies by inhibiting immune checkpoint molecules with anti- PD- 1/ ria to evade host adaptive immune responses is to predominantly PD- L1 antibodies, PD1/PDL1- directed experimental therapies in in infect macrophages instead of dendritic cells, the former requiring vitro and in vivo TB models resulted in impaired intracellular control stronger activation before being able to efficiently process and pre- of Mtb and TB exacerbation rather than improved resolution,126,127 sent antigens for priming naive T- cells.119 Macrophage activation is suggesting PD- 1 may be a T- cell activation rather than exhaustion required to induce expression of CIITA, a major positive regulator of marker during TB. Moreover, reports of LTBI- reactivation in cancer KILINÇ et aL | 11 patients treated with anti- PD- 1/PD- L1128 warrant a cautionary note hallmark Th2- cytokine, impaired mycobacterial control by human against this therapy in TB. macrophages and enhanced the proportion of regulatory T- cells in Indoleamine 2,3- dioxygenase (IDO) expression is actively in- vitro.131 Blocking IL- 4 completely alleviated these effects and im- duced by mycobacteria in animal (macaques and mice) models of proved bacterial control,131 suggesting Th skewing could be an inter- acute TB, but not LTBI, and IDO levels correlated with bacterial esting target for HDT. burden. IDO catabolizes tryptophan into kynurenine, which in Alternatively, administration of IL- 2, which stimulates T- cell pro- turn suppresses IFN-γ production by CD4 T- cells, a cytokine piv- liferation while inhibiting T- cell anergy, in patients infected with otal in the anti- TB response, identifying IDO as potential target drug- resistant Mtb has been investigated in five RCTs and compared for HDT. In vivo inhibition of IDO activity using D- 1MT one week in a meta- analysis.146 While CD4 T- cell numbers increased and time after mycobacterial infection enhanced T- cell proliferation and to culture conversion improved, radiographic changes were not ob- differentiation in effector and memory cells while apoptosis was served compared to standard chemotherapy.146 In mice infected enhanced.129 Furthermore, D- 1MT treatment improved penetra- with Mav, IL- 2 therapy resulted in decreased bacterial burden,147 tion of T- cells into granulomas, likely allowing protective T- cell- whereas mixed results were described in case reports.148,149 The lim- mediated granuloma reorganization, and reduced bacterial loads ited effect of IL- 2 therapy may be due to immune suppression caused and lung pathology.129 by expansion of regulatory T- cells and myeloid- derived suppressor cells (MDSC), both expressing elevated levels of the high affinity IL- 2 receptor, as depletion of these suppressor cells improved outcome 3.2 | Skewing of T- cells in a mouse TB model.150 Combining IL- 2 therapy with mycobacterial phosphoantigen (E)- 4- hydroxy- 3- methyl- but- 2- enyl pyrophosphate Th1- responses, characterized by high IFN-γ secretion, are cru- (HMBPP) in non- human primates induced significant expansion of 130- 132 cial in effective anti- Mtb immune responses. Nevertheless, Vγ2Vδ2 T- cells that migrated to the lungs, evoking a Th1- response Mav and Mtb reduce cellular responses to IFN-γ and deficiencies that significantly reduced mycobacterial burden as well as lung pa- 151 in the IL- 12/IL- 23/IFN-γ - axis increase susceptibility to Mav infec- thology. Rather than systemic administration of cytokines, which tions.133,134 In several patients suffering from pulmonary TB, direct frequently results in systemic side effects, ex vivo stimulation of au- administration of IFN-γ accelerated sputum- smear conversion and tologous peripheral blood mononuclear cells (PBMCs) with a cocktail 135,136 improved chest radiograph. Administration of IFN-γ also re- of IFN-γ , IL- 2, IL- 1α, and anti- CD3 before reinfusion yielded positive duced Mav growth in murine macrophages,137 and improved clini- results with minimal side effects in a case report with MDR- TB.152 cal outcome (ie, decreased respiratory symptoms and mortality) in This, however, requires further clinical investigation. several but not all Mav- infected individuals,138- 140 suggesting po- tential of IFN-γ as HDT in both Mtb and Mav infections. In vivo administration of IL- 12, a key cytokine that drives Th1 skewing, en- 4 | (PROGRAMMED) CELL DEATH hanced IFN-γ and TNF-α responses and significantly reduced bac- terial burden in an acute mouse TB model.141 Similarly, restoring Several types of cell death can follow mycobacterial infection of IL- 24 expression in a mouse TB model enhanced Th1- responses and macrophages: apoptosis, necrosis, and ferroptosis.32 During apop- IFN-γ production, with concomitant improved survival and reduced tosis, bacteria remain encapsulated, which facilitates bacterial clear- bacterial loads.142 A large proportion of human Mtb- specific CD4 ance; however, pathogenic mycobacteria have developed strategies 153 Th1- cells expresses CCR6 and co- produces IFN-γ /IL- 17, often de- to limit apoptosis. Activation of transcriptional regulator peroxi- picted as Th1* or Th1- 17 cells, and being associated with LTBI sug- some proliferator- activated receptor gamma (PPARγ) by ManLAM, gest their importance in protection against active TB.143 However, stimulating mannose receptors, upregulated (pro- host- cell survival) IL- 17 responses during TB need to be carefully regulated to prevent Mcl- 1 and repressed (pro- apoptotic) Bax without Bak and improved 154,155 neutrophil- driven lung pathology, which is mediated by regulatory host- cell survival. In agreement with the PPARγ- dependent in- T- cells as well as so- called regulatory CD4 Th17- cells that co- hibition of host- cell apoptosis and concomitant anti- mycobacterial produce IL- 17 and IL- 10.144 In case of disbalanced Th17 responses immunity, direct pharmacological inhibition of Mcl- 1 resulted in re- with concomitant excessive neutrophil recruitment, RAGE recep- duced intracellular Mtb growth in human macrophages.155 tor inhibition may be an interesting HDT. RAGE receptor is up- Besides inhibiting apoptosis, virulent Mtb stimulates host- cell ne- regulated during active TB disease and after ligation with S100A8/ crosis, which allows infection to disseminate to neighboring cells.32 A9 mediates neutrophil recruitment.145 In a TB model, mice defi- Mtb can induce necrosis via the virulence factor tuberculosis necro- cient in S100A8/A9 had reduced bacterial loads, neutrophil influx tizing toxin (TNT), which is secreted into the cytosol where its NAD+ and pathology compared to wildtype. Moreover, inhibition of the glycohydrolase activity depletes the host cell from NAD+,156 leading RAGE receptor using FPS- ZM1 improved outcome comparably as to permeabilization of mitochondrial membranes, decreasing ATP- S100A8/A9- deficiency.145 production and activating necrosis. Nicotinamide- based HDT allevi- Th2- responses have been associated with active cavitary TB ated necrosis- induced host- cell cytotoxicity in Mtb- infected cells by disease or TB treatment failure,130,131 and administration of IL- 4, a replenishing NAD+.99 Mtb can furthermore induce necrosis mediated by 12 | KILINÇ et aL mitochondrial membrane permeability transition via p38- MAPK phos- a result of ATP depletion induced macrophage apoptosis.174 An impor- phorylation, which can be inhibited by corticosteroids dexamethasone tant enzyme during glycolysis, lactate dehydrogenase (LDH), which and doramapimod.157 In addition, corticosteroids dexamethasone and converts pyruvate into lactate, is significantly upregulated during Mtb prednisolone, both well- known general immunosuppressants, have infection.175 Although the pathophysiology of LDH upregulation re- also been investigated as HDT during mycobacterial infections. With mains to be addressed, pharmacological inhibition of LDH using FX11 some reports of improved survival,158,159 likely by limiting secretion reduced bacterial load and development of necrotic lesions in granu- of pro- inflammatory cytokines, meta- analysis failed to show a signif- lomas in a murine TB model, suggesting a significant role of LDH in icant improvement in clinical outcome after corticosteroid therapy in driving disease and a potential target for HDT.175 Interestingly, while patients with TB.160 Interestingly, while promoting pro- inflammatory ATP depletion can induce macrophage apoptosis (considered host pro- cytokine levels of TNF-α by adenylate cyclase inhibitor (SQ22536) or tective), exogenous ATP activates macrophages via the P2RX7/P2X7 a PKA inhibitor (H- 89) has been shown to improve control of infection receptor and also directly inhibits growth of mycobacteria, including 161 176,177 by stimulating mitochondrial ROS production, excess TNF-α leads Mtb and Mav, due to chelation of iron. ATP treatment has already to membrane disruption and ATP depletion via mitochondrial enzyme been shown to synergize with standard Mav antibiotic treatment, mak- cyclophilin D, which together with lysosomal enzyme acid sphingo- ing ATP an interesting adjunctive HDT molecule to enhance chemo- myelinase induced necrosis.162,163 Alisporivir and desipramine, two therapeutic efficacy against mycobacterial infections.177 In addition, clinically approved drugs that inhibit cyclophilin D and acid sphingomy- an FDA- approved potentiator of P2RX7/P2X7, clemastine enhanced 178 elinase, respectively, prevented TNF-α - induced necrosis without com- mycobacterial killing in a zebrafish model. This may provide a poten- 163 promising TNF-α - induced ROS- dependent mycobacterial killing. tially attractive avenue to explore synergistic effects between ATP and Correspondingly, upregulation of cAMP levels by phosphodiesterase clemastine treatment in future studies. (PDE) inhibitors cilostazol and sildenafil decreased TNF-α levels, result- Conversion of pyruvate into acetyl coenzyme A (Ac- CoA) ini- ing in reduced immunopathology and fastened bacterial clearance in tiates the tricarboxylic acid (TCA) cycle which produces energy 164,165 Mtb- infected mice. Blocking TNF- α, which facilitates necrotizing using OXPHOS. During Mtb infection, several enzymes important granulomas during active TB, displayed promising results in preclinical in the TCA cycle are downregulated and TCA cycle intermediates, 166,167 animal models. However, blocking TNF-α also leads to disease such as citrate, are translocated from mitochondria into the cyto- reactivation and concomitant dissemination in LTBI patients and in the sol. Typically, citrate is converted into itaconate which dampens absence of standard TB chemotherapy exacerbated disease severi- tissue hyperinflammation by suppressing both ROS production and 168- 170 ty, precluding clinical application of TNF-α inhibition as HDT in production of pro- inflammatory cytokines such as IL- 1β, IL- 6, and IL- 174 TB. The balance between TNF-α - mediated beneficial and detrimental 12. In the cytosol, however, citrate is cleaved into Ac- CoA, which effects on host control of TB and likely other mycobacterial infections is either converted into arachidonic acid or into mevalonate and including NTM is thus delicate. Taken together, these data indicate that malonyl- CoA. This leads to synthesis of eicosanoids, cholesterol, Mtb- induced host- cell necrosis favors mycobacterial survival and this and free fatty acids, respectively, of which the latter two are stored can be effectively counteracted by HDT, while the double- edge sword intracellularly in lipid droplets.174 Hypercholesterolemia results in of modulating TNF-α levels currently prohibits clinical application. spontaneous formation of lipid droplets in macrophages. Further Ferroptosis is a type of necrosis characterized by accumulation accumulation of intracellular lipid droplets is actively stimulated by 171 174,179 of free iron and toxic lipid peroxides. In Mtb- infected cells expres- Mtb and enhanced by both IL- 6 and TNF-α signaling, while IL- 174 sion of glutathione peroxidase- 4 (GPX4) is reduced, leading to failure 17 and IFN-γ limit intracellular lipid accumulation. Lipid- loaded of glutathione- dependent antioxidant defenses and cell death.172 macrophages are impaired in killing intracellular mycobacteria (ie, Inhibiting ferroptosis by ferrostatin 1 reduced bacterial burden both Mtb, Mav, and BCG)180 and ultimately transform into foamy macro- in vitro in human macrophages and in vivo in Mtb- infected mice.171 phages, which are associated with necrotic granulomas and tissue Furthermore, ferroptosis could also be inhibited by increasing GPX4 pathology.174 The impaired functionality of lipid- loaded macro- levels with selenium, a protein involved in GPX4 catalysis,173 show- phages involves mitochondrial dysfunction and could be restored ing that targeting this host pathway is a potential HDT strategy. using small molecule mitochondrial fusion promoter M1, which also restored macrophage bactericidal activity.180 In addition, eze- timibe, a cholesterol absorption inhibitor, prevented intracellular 5 | METABOLISM lipid accumulation and concomitantly reduced intracellular growth in Mtb- infected macrophages.179 The effects of standard antibiotic 5.1 | Carbohydrate and lipids treatment improved and perhaps even synergized with ezetimibe treatment,179 and investigating the in vivo efficacy of ezetimibe as Mtb has developed numerous strategies to modulate host metabolic well as M1 could be promising. pathways, which are broadly divided into glycolysis, oxidative phos- Statins, currently clinically used to reduce cholesterol levels, phorylation (OXPHOS), and lipid metabolism. Glycolysis conditions an could be interesting drugs to prevent lipid accumulation in macro- environment favoring Mtb growth, and inhibition of glycolysis using phages. Comparing eight different statins, simvastatin, pravastatin 2- deoxy- D- glucose (2- DG) reduced Mtb viability in one study, and as and fluvastatin were most efficacious in enhancing mycobacterial KILINÇ et aL | 13

181 killing without affecting cell viability in vitro. Mechanistically, mycobacterial infection due to absence of LTB4. Treatment with ce- while (simva)statin inhibits phagosomal acidification and degrada- lecoxib, a COX inhibitor that prevents PGE2 production and thereby 181 tion, cholesterol incorporation in (auto- )phagosomal membranes stimulates LTB4 production, or directly supplementing LTB4, re- is prevented. The presence of cholesterol in phagosomal membranes stored mycobacterial control.187 Furthermore, COX inhibitors ibu- facilitates prolonged survival of Mtb and Mav within host cells due profen and aspirin administered as single therapy or combined with to blockage of phagolysosome fusion by mechanisms not fully un- conventional TB antibiotics were shown to limit bacterial burden in derstood.182- 184 Preventing phagosomal escape ultimately enhances Mtb- infected mice,192- 194 and low- dose aspirin treatment also re- delivery of mycobacteria to (auto- )phagolysosomes and thereby duced bacterial loads in a Mm zebrafish infection model.195 Aspirin bacterial degradation.185,186 In vivo treatment with either pravasta- treatment of TB or TB meningitis patients improved survival,196,197 tin or simvastatin in a mouse TB model reduced mycobacterial loads but may impair conventional treatment regimens by reducing effi- both as a single therapy181,185 or combined with standard antibiotic cacy of isoniazid,198 but not pyrazinamide.193 Both ibuprofen and treatment.181,184 aspirin are currently tested in clinical trials as adjunct therapy for treating (drug- resistant) TB.190

5.2 | Eicosanoids 5.3 | GRANULOMA: FORMATION, Eicosanoids are lipid mediators involved in regulating inflammatory ANGIOGENESIS and HYPOXIA responses and are categorized into prostaglandins (PG), leukotrienes (LT), thromboxanes, lipoxins, and hydroxy eicosatetraenoic acids, One hallmark of TB is the extensive formation of granulomas. all of which are produced from arachidonic acid by a competing Granulomas are highly heterogenous and dynamic structures which network of enzymes, including cyclooxygenases (COX) and lipoxy- differ significantly in the level of hypoxia and available nutrients. genases.187,188 During Mtb infection, the expression of eicosanoids Granuloma formation is actively initiated by Mtb to stimulate ma- is significantly altered, with prostaglandin- E2 (PGE2) and leukot- trix metalloprotease 9 (MMP9) production. Granulomas are also 187 199,200 201 riene- B4 (LTB4) mostly upregulated. Being an immune suppres- induced during NTM infections, including Mav and Mm. sor and immune stimulator, respectively, the balance between these During initial granuloma formation non- activated macrophages are eicosanoids is highly important in regulating immunity to clear the recruited to the site of infection and serve as feeder cells for the infection, without causing tissue pathology due to excessive in- granuloma.25,202 In addition to MMP9, upregulation of several other flammation. Important in this regulation are IL- 1β and type- I IFN MMPs has been observed in lung samples from individuals infected signaling. IL- 1β signaling stimulates production of prostaglandin- with Mtb, and other mycobacteria including Mav, which may suggest E2, which is necessary to dampen the mediated by that similar mechanisms are involved.202- 206 MMPs are enzymes that pro- inflammatory leukotrienes A4 and B4 (LTA4 is the precursor of degrade and modulate extracellular matrix and are therefore key in 204 LTB 4) that are induced upon type- I IFN signaling. In severe TB, the the development of granulomas. Their expression and activity 2+ PGE2/LTA4 ratio is reduced, suggesting potential benefit of enhanc- has multiple layers of regulation. Many MMPs require Zn for their ing PGE2 signaling. Indeed, both increasing PGE2 levels using ad- activation, potent MMPs require activation by other MMPs, and ministration of exogenous PGE2 or reducing LTA4/LTB4 production their activation is inhibited by tissue inhibitors of metalloproteinases with zileuton improved host survival, while reducing bacterial loads (TIMPs). Expression of MMPs is stimulated by pro- inflammatory 189 and necrotic lung pathology in Mtb- infected mice. Moreover, cytokines, including IFN-γ , TNF-α , IL- 12, and IL- 17 and because en- combinatory therapy of zileuton with PGE2 further restricted Mtb hanced MMP- activity is associated with extensive tissue damage replication.190 during TB,203 MMPs are promising targets for HDTs. A single nucleotide polymorphism in the promotor of the gene MMP1, a collagenase that degrades collagen in the extracellular encoding LTA4- hydrolase (rs17525495), the enzyme that converts matrix, is upregulated after TLR2- ligation and due to its high potency 207 LTA4 into LTB4, has been shown to affect expression of LTA4 hydro- may drive granuloma formation during TB. In transgenic mice ex- lase, with homozygous individuals having a high (T/T) or low (C/C) pressing human MMP1, Mtb infection promoted alveolar destruc- expression.188,191 Both homozygous genotypes have poorer survival tion and collagen breakdown in lung granulomas, identifying MMP1 compared to heterozygous individuals, showing the delicateness of as a therapeutic target to limit immunopathology.207 MMP7, which is the immune balance during mycobacterial infection.188 Depending highly expressed in the cavitary wall and hypoxic granulomas, stim- on the genotype, different treatment regimens will be required, as ulates epithelial proliferation and promotes activity of other MMPs. general immune suppression using dexamethasone favored outcome Inhibition of MMP7 and MMP1 using cipemastat, a drug originally in T/T individuals, while being detrimental in C/C individuals,188,191 registered to prevent lung fibrosis, surprisingly increased cavita- suggesting the necessity of personalized HDT- based medicine tar- tion, immunopathology and mortality in mice,208 suggesting either geting eicosanoid metabolism. Mice deficient in 5- lipoxygenase, an a protective role of MMP1 or MMP7 during TB or off- target effects enzyme that stimulates production of LTA4 and thus LTB4 (thereby of the drug. The role of MMP8 is more controversial with high in- being a model for C/C individuals), were impaired in controlling terindividual variation,203,209,210 which may relate to the presence 14 | KILINÇ et aL of neutrophils in granulomas. MMP8 is more readily detectable in host cells to stimulate angiogenesis.216 Although angiogenesis could HIV- associated TB,210 suggesting that neutrophils are recruited potentially increase host- cell viability, the net effect likely favors preferentially in settings of impaired adaptive immunity. Mice defi- bacterial replication and dissemination. Blocking angiogenesis may cient in MMP9 have less granuloma formation and reduced bacterial therefore be an interesting HDT. Indeed, inhibition of VEGF using loads,211 suggesting a prominent role of MMP9 in driving disease pa- FDA- approved bevacizumab in Mtb- infected rabbits, reduced the thology. Indeed, inhibition of MMP9 expression using morpholinos total number of vessels but improved both structurally and func- reduced granuloma formation and bacterial growth in a zebrafish tionally the remaining vessels, leading to enhanced drug targeting Mm- model.25 In agreement with this concept, treatment with Sb- 3ct, to granulomatous lesions and diminished hypoxia.217 Corroborating a specific MMP2 and MMP9 inhibitor, combined with frontline TB these findings, treatment of Mm- infected zebrafish with VEGF path- antibiotics potentiated bacterial clearance both in vitro and in vivo way inhibitors SU5416, a tyrosine kinase inhibitor, or pazopanib, a in a TB meningitis mouse model.212,213 Blocking MMP9 using mono- VEGF receptor inhibitor, reduced bacterial loads and dissemination. clonal antibody AB0046 did not affect bacterial burden, but the rate Both drugs also synergized with first- line anti- mycobacterial drugs of relapse was reduced in a necrotic granuloma TB mouse model, rifampicin and metronidazole, a drug that targets hypoxic bacte- by mechanisms not yet fully clarified.166 Using an in vitro model for ria.218 Inhibiting vascular leakage rather than angiogenesis may be extracellular matrix degradation, treatment with doxycycline, an equally efficacious to limit nutrient supply to mycobacteria. During FDA- approved antibiotic that non- selectively inhibits human MMPs, Mm infection, angiopoietin- 2 (ANG2) is robustly induced in granu- strongly abolished Mtb- induced matrix degradation.210 In addition, lomatous lesions. ANG2 antagonizes ANG1, which promotes vessel doxycycline reduced granuloma formation in a guinea pig model, stability while limiting angiogenesis and vascular leakage. Indeed, likely resulting from abolishing Mtb- enhanced promotor activity of AKB- 9785, a molecule that mimics functions of ANG1, reduced MMP1 and by directly inhibiting bacterial growth.214 Pan-MMP in- vascular leakage and bacterial burden in a Mm zebrafish infection hibitor marimastat (BB- 2516), a collagen- peptidomimetic binding the model.219 Thus, inhibition of angiogenesis is an interesting target for active Zn2+ site contained in many MMPs, reduced granuloma size HDT to enhance drug delivery to the site of infection and combined and bacterial burden during Mtb infection in lung tissue models.203 with other therapies is likely to be even more potent. Interestingly, treatment of Mtb- infected mice with a panel of MMP inhibitors, including marimastat, as solo therapy was not effective, while all 4 small molecules enhanced in vivo potency of frontline TB 6 | PERSONALIZED AND drugs isoniazide and rifampicin, likely by blocking MMP- mediated COMBINATORIAL HDT cleavage of collagen and by improving vascular integrity, resulting in enhanced delivery of isoniazide and rifampicin to the lungs. The Although HDT could be considered as stand- alone therapy, for ex- finding that batimastat (a pan- MMP inhibitor), Sb- 3ct (a MMP2 ample, in patients suffering from total drug- resistant TB, HDT is and MMP9 inhibitor) and MMP9 inhibitor- I yielded similar results primarily envisaged as adjunct therapy in combination with classical highlights the importance of MMP9 in driving these effects.205 antibiotics. HDT might be co- administered for a limited duration at Augmenting TIMP1 activity to inhibit activity of multiple MMPs may the initiation of the standard of care regimens to shorten treatment also be an interesting HDT target. To our knowledge, however, mod- length and reduce dosage of antibiotics to minimize side effects, ulating the activity of TIMPs has not been investigated yet in the or toward the end of treatment to boost host immunity to prevent context of HDT. potential relapse. Consequently, investigating the interactions be- Central hypoxia in granulomas may initially favor host immunity tween HDT and conventional chemotherapy is pivotal, but has only as low oxygen tension increases granulysin expression in T- cells and been reported for a limited number of HDTs. Furthermore, in case NK- cells, enhancing bacterial killing in an in vitro co- culture system of undesired interactions between TB drugs and drugs for TB co- of Mtb- specific T- cells and macrophages.215 However, due to poor morbidities (eg, between rifampicin and anti- HIV therapy or anti- 220 vasculature within granulomas and hyperactive IFN-γ or possible diabetic drugs), HDT might be used to shorten current treatment superimposed IL- 4/IL- 13 released by activated T- cells, full blown regimens or possibly partially replace components of the conven- central necrosis leads to cavity formation and concomitant bac- tional chemotherapy cocktail. In line with this, interactions between terial dissemination within the host.202,211,215 Due to the hypoxic, HDT and drugs used to treat TB comorbidities should also be inves- acidic and nutrient- poor conditions in granulomas, mycobacterial tigated thoroughly. dormancy is promoted,25 and while this effectively inhibits bac- Rather than targeting one specific aspect of the inflammatory terial replication, eradication of mycobacteria is greatly hampered response during mycobacterial infections, we hypothesize that cor- because most antibiotics only affect replicating and metabolically recting the overall immunological disbalance likely is most promising. active bacteria. Furthermore, poor vascularization hampers drug de- Type- I IFN and IL- 1β signaling, regulating levels of anti- inflammatory livery in granulomas, which is further impaired due to fibrosis and prostaglandins and pro- inflammatory leukotrienes, respectively, play scarring of lung tissue caused by the disease.25 Trehalose dimyco- an important role in regulating the immune balance during mycobac- late, a mycolic acid expressed on mycobacterial cell walls, directly terial infections.189 At the time of writing, multiple randomized con- induces vascular endothelial growth factor (VEGF) expression in trolled trials investigate targeting of (one of) these pathways by HDTs. KILINÇ et aL | 15

As some TB patients suffer from overactive type- I IFN/leukotriene mycobacteria. Targeting metabolism with HDT may also help cor- signaling while others are characterized by overactive IL- 1/prostaglan- recting the balance between prostaglandins and thromboxanes, as din activity, we postulate that in this context personalized HDT would lipid droplets and cytosolic TCA intermediates are the most import- be safest and most efficacious. However, this will increase therapeu- ant sources of eicosanoids. tic costs, which could make such therapy stratifications less attractive Irrespective of what causes defective mycobacterial clearance, and feasible in lower resourced settings. To be able to predict whether improving drug delivery to the site of infection likely benefits all patients would benefit from a certain HDT, biomarkers monitoring TB patients. Angiogenesis in granulomas is significantly impaired the (immunological) status of patients may need to be identified and and further enhances hypoxia and nutrient- limitation. Targeting developed. This, however, may not be required for all HDTs as some angiogenesis during TB by (a) inhibiting VEGF (bevacizumab217), (b) HDT may improve anti- mycobacterial immunity in all patients. As inhibiting VEGF- mediated signaling (SU5416 or pazopanib218), or (c) mycobacteria modulate host immunity via many different pathways, antagonizing pro- angiogenesis growth factor ANG2 (AKB- 9785219) a multi- targeted approach could be necessary to fully counteract has all been shown to enhance both drug delivery and oxygenation mycobacteria- mediated host modulation. To our knowledge, however, within granulomas in animal models of TB, and may be promising only two combinations of HDT treatments have been published; com- HDTs in combination with other therapies. Despite being most fre- bining vitamin- D with PBA did not mediate additive effects compared quently investigated in combination with antibiotics, efflux pump to solo therapy, 51- 53 likely because both compounds target the same inhibitors could also improve drug delivery of HDTs. To our knowl- pathway, while in another in vitro study combining protein- kinase A/B edge, however, this has not been investigated so far but verapamil, inhibitors H- 89 or 97i with HDAC inhibitors revealed additive effects known to enhance the efficacy of rifampicin and bedaquiline against in vitro in reducing bacterial load in primary human macrophages.115 different mycobacterial infections, both in vitro and in mice,222- 224 Modulating (auto- )phagosome maturation using receptor tyro- and also chloroquine225 and piperine 226 are interesting molecules sine kinase inhibitors including imatinib,25 AZD0530,28 and multiple for combinatorial HDT. repurposed drugs recently identified in our own group29 has been Given their central and important role in orchestrating a func- shown to improve mycobacterial clearance by human macrophages tional anti- mycobacterial immune response, restoring (CD4 Th1/17) in vitro. Importantly, releasing the mycobacteria- mediated arrest in T- cell immunity has been pursued in many investigations. In addi- (auto- )phagosome maturation likely benefits both patients with ac- tion to enhancing activation of antigen- presenting cells, HDTs that tive disease as well as individuals with latent infection. Above, we promote phagosomal bacterial degradation (ie, stimulating auto- have reviewed multiple HDT candidates that enhance autophagy- phagy, enhancing phagosome maturation and promoting (auto- ) mediated bacterial clearance. Which of these will be most efficacious phagolysosome fusion) are all expected to enhance presentation against mycobacteria should ideally be determined in head- to- head of bacterial- derived peptides and thereby improve adaptive immu- comparisons. Metformin, being the most frequently investigated, nity. Modulating T- cell responses to restore immunity can be medi- has already been shown to reduce TB recurrence and bacterial loads ated by vaccination or T- cell cytokine therapies. Administration of in patients,75- 77 and in addition to its effects on autophagy, also en- IL- 12141 or IL- 24142 or blocking Th2 cytokine IL- 4,131 promotes Th1 221 hances mitochondrial membrane polarization, which could fur- responses with lasting IFN-γ production that may be preferred over ther enhance its efficacy. IFN-γ administration. Which of these strategies is (most) efficacious As discussed above, host- cell death pathways are actively ex- and which patients benefit most from this therapy remains to be ploited by mycobacteria to promote their survival and dissemination addressed. and have been shown to be a potential target for HDT in multiple in While most of the evidence available for host- pathogen inter- vitro and animal studies. Active clinical modulation of (programmed) actions and HDT are from TB studies, the limited number of NTM cell death in patients, however, could lead to significant adverse experimental models investigating host modulation and/or HDT effects given the complex time- and context- dependency of this emphasizes the need and urgency to understand NTM pathogen- mechanism during mycobacterial infection. esis as well as identify potentially relevant host targets. Together, Targeting metabolic pathways has been shown to be feasible these studies will help assess the safety and efficacy of HDT, pav- and represents an attractive target for HDT. While most metabolic ing the way for the introduction of HDT against a wide range of pathways are also necessary for host- cell energy production, intra- mycobacteria. cellular lipid accumulation in lipid droplets seems to mainly benefit the intracellular survival of mycobacteria. Preventing or reducing lipid droplet formation in macrophages and concomitant impaired 7 | SEARCH STRATEGY AND SELECTION immunity can be mediated by (a) limiting oxidative phosphorylation CRITERIA by, for example, stimulating polarization of macrophages toward pro- inflammatory M1- macrophages,174 (b) improving/maintaining We searched PubMed (MEDLINE) for all relevant studies pub- mitochondrial membrane potential using small molecule M1180 or lished from 1 January 2000 until 1 October 2020. The medical NAD156 and/or (c) blocking cellular cholesterol uptake using, for subject headings used were “host directed”, “HDT”, “adjunctive”, example, ezetimibe,179 which also inhibits phagosomal escape by “immunotherapeutic” or “immunomodulation” combined with 16 | KILINÇ et aL

10. Rhoades ER, Archambault AS, Greendyke R, Hsu FF, Streeter C, “mycobacterium”, “mycobacteria”, “tuberculosis”, “nontuberculous” Byrd TF. Mycobacterium abscessus Glycopeptidolipids mask un- or “NTM”. All relevant abstracts were screened independently by derlying cell wall phosphatidyl- myo- inositol mannosides blocking two researchers. The final reference list was generated based on rel- induction of human macrophage TNF- alpha by preventing interac- evance to the topics covered in this review. Only papers published in tion with TLR2. J Immunol. 2009;183(3):1997- 2007. 11. Rahman A, Sobia P, Gupta N, Kaer LV, Das G. Mycobacterium tu- English were included. berculosis subverts the TLR- 2- MyD88 pathway to facilitate its translocation into the cytosol. PLoS One. 2014;9(1):e86886. ACKNOWLEDGEMENTS 12. Gilleron M, Nigou J, Nicolle D, Quesniaux V, Puzo G. The acylation This work was supported by grants from the Netherlands state of mycobacterial lipomannans modulates innate immunity response through toll- like receptor 2. Chem Biol. 2006;13(1):39- 47. Organisation for Scientific Research (NWO- TTW) within the Novel 13. Khan N, Pahari S, Vidyarthi A, Aqdas M, Agrewala JN. Stimulation Antibacterial Compounds and Therapies Antagonising Resistance through CD40 and TLR- 4 is an effective host directed therapy (NACTAR) program (Grant No. 16444), European Union's Horizon against Mycobacterium tuberculosis. Front Immunol. 2016;7:386. 2020 research and innovation program (SMA- TB Grant No. 847762), 14. Khan N, Pahari S, Vidyarthi A, Aqdas M, Agrewala JN. NOD- 2 and TLR- 4 signaling reinforces the efficacy of dendritic cells and and the Innovative Medicines Initiative 2 Joint Undertaking (IMI2 reduces the dose of TB drugs against Mycobacterium tuberculosis. JU) under the RespiriNTM (Grant No. 853932) and RespiriTB (Grant J nnate Immun. 2016;8(3):228- 242. No. 853903) projects within the IMI AntiMicrobial Resistance (AMR) 15. Gowthaman U, Singh V, Zeng W, et al. Promiscuous peptide of 16 Accelerator program. The funders had no role in study design, data kDa antigen linked to Pam2Cys protects against Mycobacterium tuberculosis by evoking enduring memory T- cell response. J Infect collection and analysis, decision to publish, or preparation of the Dis. 2011;204(9):1328- 1338. manuscript. 16. Chodisetti SB, Gowthaman U, Rai PK, Vidyarthi A, Khan N, Agrewala JN. Triggering through Toll- like receptor 2 limits chron- CONFLICT OF INTEREST ically stimulated T- helper type 1 cells from undergoing exhaustion. J Infect Dis. 2015;211(3):486- 496. All authors declare no competing or conflicting interests. 17. Pahari S, Negi S, Aqdas M, Arnett E, Schlesinger LS, Agrewala JN. Induction of autophagy through CLEC4E in combination with TLR4: DATA AVAILABILITY STATEMENT an innovative strategy to restrict the survival of Mycobacterium tu- Data sharing not applicable to this article as no datasets were gener- berculosis. Autophagy. 2020;16(6):1021- 1043. ated or analysed during the current study 18. Harding CV, Boom WH. Regulation of antigen presentation by Mycobacterium tuberculosis: a role for Toll- like receptors. Nat Rev Microbiol. 2010;8(4):296- 307. ORCID 19. Kim JK, Yuk JM, Kim SY, et al. MicroRNA- 125a inhibits autophagy Tom H. M. Ottenhoff https://orcid.org/0000-0003-3706-3403 activation and antimicrobial responses during mycobacterial infec- Mariëlle C. Haks https://orcid.org/0000-0002-7538-1800 tion. J Immunol. 2015;194(11):5355- 5365. 20. Gu X, Gao Y, Mu DG, Fu EQ. MiR- 23a- 5p modulates mycobacte- rial survival and autophagy during mycobacterium tuberculosis REFERENCES infection through TLR2/MyD88/NF- kappaB pathway by targeting 1. World Health Organization. Tuberculosis. https://www.who.int/ TLR2. Exp Cell Res. 2017;354(2):71- 77. news- room/fact- sheet s/detai l/tuber culosis. 2020. 21. Jefferies KC, Cipriano DJ, Forgac M. Function, structure and 2. Cilloni L, Fu H, Vesga JF, et al. The potential impact of the regulation of the vacuolar (H+)- ATPases. Arch Biochem Biophys. COVID- 19 pandemic on the tuberculosis epidemic a modelling 2008;476(1):33- 42. analysis. EClinicalMedicine. 2020;28:100603. 22. Lawrence RE, Zoncu R. The lysosome as a cellular centre 3. Wu ML, Aziz DB, Dartois V, Dick T. NTM drug discov- for signalling, metabolism and quality control. Nat Cell Biol. ery: status, gaps and the way forward. Drug Discov Today. 2019;21(2):133- 142. 2018;23(8):1502- 1519. 23. Ankley L, Thomas S, Olive AJ. Fighting persistence: how chronic 4. Schafer G, Jacobs M, Wilkinson RJ, Brown GD. Non- opsonic rec- infections with Mycobacterium tuberculosis Evade T cell- mediated ognition of Mycobacterium tuberculosis by phagocytes. J Innate clearance and new strategies to defeat them. Infect Immun. Immun. 2009;1(3):231- 243. 2020;88(7):e00916- 00919. 5. Kawai T, Akira S. TLR signaling. Cell Death Differ. 24. Wong D, Bach H, Sun J, Hmama Z, Av- Gay Y. Mycobacterium 2006;13(5):816- 825. tuberculosis protein tyrosine phosphatase (PtpA) excludes host 6. Gomes MS, Florido M, Cordeiro JV, et al. Limited role of the Toll- vacuolar-H+- ATPase to inhibit phagosome acidification. Proc Natl like receptor- 2 in resistance to Mycobacterium avium. Immunology. Acad Sci U S A. 2011;108(48):19371- 19376. 2004;111(2):179- 185. 25. Hawn TR, Matheson AI, Maley SN, Vandal O. Host- directed ther- 7. Drennan MB, Nicolle D, Quesniaux VJ, et al. Toll- like receptor apeutics for tuberculosis: can we harness the host? Microbiol Mol 2- deficient mice succumb to Mycobacterium tuberculosis infec- Biol Rev. 2013;77(4):608- 627. tion. Am J Pathol. 2004;164(1):49- 57. 26. Napier RJ, Rafi W, Cheruvu M, et al. Imatinib- sensitive tyro- 8. Orme IM, Ordway DJ. Host response to nontuberculous myco- sine kinases regulate mycobacterial pathogenesis and repre- bacterial infections of current clinical importance. Infect Immun. sent therapeutic targets against tuberculosis. Cell Host Microbe. 2014;82(9):3516- 3522. 2011;10(5):475- 485. 9. Bhatnagar S, Schorey JS. Elevated mitogen- activated protein ki- 27. Bruns H, Stegelmann F, Fabri M, et al. Abelson tyrosine ki- nase signalling and increased macrophage activation in cells in- nase controls phagosomal acidification required for killing of fected with a glycopeptidolipid- deficient Mycobacterium avium. Mycobacterium tuberculosis in human macrophages. J Immunol. Cell Microbiol. 2006;8(1):85- 96. 2012;189(8):4069- 4078. KILINÇ et aL | 17

28. Chandra P, Rajmani RS, Verma G, Bhavesh NS, Kumar D, 48. Huang SJ, Wang XH, Liu ZD, et al. Vitamin D deficiency and Stallings CL. Targeting drug- sensitive and - resistant strains of the risk of tuberculosis: a meta- analysis. Drug Des Dev Ther. Mycobacterium tuberculosis by Inhibition of Src family kinases low- 2017;11:91- 102. ers disease burden and pathology. mSphere.. 2016;1(2):e00043 49. Coussens AK, Wilkinson RJ, Martineau AR. Phenylbutyrate - e15. is bacteriostatic against Mycobacterium tuberculosis and regu- 29. Korbee CJ, Heemskerk MT, Kocev D, et al. Combined chemical ge- lates the macrophage response to infection, synergistically with netics and data- driven bioinformatics approach identifies recep- 25- hydroxy- vitamin D3. PLoS Pathog. 2015;11(7):e1005007. tor tyrosine kinase inhibitors as host-directed antimicrobials. Nat 50. Rekha RS, Muvva SSVJR, Wan M, et al. Phenylbutyrate in- Commun. 2018;9(1):358. duces LL- 37- dependent autophagy and intracellular killing of 30. Buter J, Cheng TY, Ghanem M, et al. Mycobacterium tuberculosis Mycobacterium tuberculosis in human macrophages. Autophagy. releases an antacid that remodels phagosomes. Nat Chem Biol. 2015;11(9):1688- 1699. 2019;15(9):889- 899. 51. Mily A, Rekha RS, Kamal SMM, et al. Oral intake of phenylbutyrate 31. Houben D, Demangel C, van Ingen J, et al. ESX- 1- mediated trans- with or without vitamin D- 3 upregulates the cathelicidin LL- 37 in location to the cytosol controls virulence of mycobacteria. Cell human macrophages: a dose finding study for treatment of tuber- Microbiol. 2012;14(8):1287- 1298. culosis. Bmc Pulm Med. 2013;13:23. 32. Chai Q, Wang L, Liu CH, Ge B. New insights into the evasion of 52. Bekele A, Gebreselassie N, Ashenafi S, et al. Daily adjunctive ther- host innate immunity by Mycobacterium tuberculosis. Cell Mol apy with vitamin D3 and phenylbutyrate supports clinical recov- Immunol. 2020;17(9):901- 913. ery from pulmonary tuberculosis: a randomized controlled trial in 33. Ranjbar S, Haridas V, Nambu A, et al. Cytoplasmic RNA sensor path- Ethiopia. J Intern Med. 2018;284(3):292- 306. ways and nitazoxanide broadly inhibit intracellular Mycobacterium 53. Mily A, Rekha RS, Kamal SM, et al. Significant effects of oral tuberculosis growth. iScience. 2019;22:299- 313. phenylbutyrate and vitamin D3 adjunctive therapy in pulmo- 34. Walsh KF, McAulay K, Lee MH, et al. Early bactericidal activity nary tuberculosis: a randomized controlled trial. PLoS One. trial of nitazoxanide for pulmonary tuberculosis. Antimicrob Agents 2015;10(9):e0138340. Chemother. 2020;64(5):e01956- e1919. 54. Tenforde MW, Yadav A, Dowdy DW, et al. Vitamin A and D defi- 35. Levine B, Mizushima N, Virgin HW. Autophagy in immunity and ciencies associated with incident tuberculosis in HIV- infected pa- inflammation. Nature. 2011;469(7330):323- 335. tients initiating antiretroviral therapy in multinational case- cohort 36. Renna M, Schaffner C, Brown K, et al. Azithromycin blocks auto- study. Jaids- J Acq Imm Def. 2017;75(3):E71- E79. phagy and may predispose cystic fibrosis patients to mycobacterial 55. Coleman MM, Basdeo SA, Coleman AM, et al. All- trans retinoic infection. Journal of Clinical Investigation. 2011;121(9):3554- 3563. acid augments autophagy during intracellular bacterial infection. 37. Paik S, Kim JK, Chung C, Jo EK. Autophagy: A new strategy for host- Am J Respir Cell Mol Biol. 2018;59(5):548- 556. directed therapy of tuberculosis. Virulence. 2019;10(1):448- 459. 56. Chen QY, Mosovsky KL, Ross AC. Retinoic acid and alpha- 38. Yuk JM, Shin DM, Lee HM, et al. Vitamin D3 induces autophagy galactosylceramide regulate the expression of costimulatory re- in human /macrophages via cathelicidin. Cell Host ceptors and transcription factors responsible for B cell activation Microbe. 2009;6(3):231- 243. and differentiation. Immunobiology. 2013;218(12):1477- 1487. 39. Liu PT, Stenger S, Li HY, et al. Toll- like receptor triggering of 57. Sada- Ovalle I, Skold M, Tian T, Besra GS, Behar SM. alpha- a vitamin D- mediated human antimicrobial response. Science. Galactosylceramide as a therapeutic agent for pulmonary mycobacte- 2006;311(5768):1770- 1773. rium tuberculosis infection. Am J Resp Crit Care. 2010;182(6):841- 847. 40. Liu PT, Stenger S, Tang DH, Modlin RL. Cutting edge: Vitamin D- 58. Mourik BC, Leenen PJ, de Knegt GJ, et al. Immunotherapy added to mediated human antimicrobial activity against Mycobacterium tu- antibiotic treatment reduces relapse of disease in a mouse model berculosis is dependent on the induction of cathelicidin. Journal of of tuberculosis. Am J Respir Cell Mol Biol. 2017;56(2):233- 241. Immunology. 2007;179(4):2060- 2063. 59. Karyadi E, West CE, Schultink W, et al. A double- blind, placebo- 41. Rockett KA, Brookes R, Udalova I, Vidal V, Hill AV, Kwiatkowski controlled study of vitamin A and zinc supplementation in persons D. 1,25- Dihydroxyvitamin D3 induces nitric oxide synthase and with tuberculosis in Indonesia: effects on clinical response and nu- suppresses growth of Mycobacterium tuberculosis in a human tritional status. Am J Clin Nutr. 2002;75(4):720- 727. macrophage- like cell line. Infect Immun. 1998;66(11):5314- 5321. 60. Visser ME, Grewal HM, Swart EC, et al. The effect of vitamin A 42. Coussens A, Timms PM, Boucher BJ, et al. 1alpha,25- and zinc supplementation on treatment outcomes in pulmo- dihydroxyvitamin D3 inhibits matrix metalloproteinases in- nary tuberculosis: a randomized controlled trial. Am J Clin Nutr. duced by Mycobacterium tuberculosis infection. Immunology. 2011;93(1):93- 100. 2009;127(4):539- 548. 61. Wang J, Xiong K, Wang Q, Zhao S, Liu Y, Ma A. Adjunctive vita- 43. Jolliffe DA, Ganmaa D, Wejse C, et al. Adjunctive vitamin D in tu- min A and D during pulmonary tuberculosis treatment: a random- berculosis treatment: meta- analysis of individual participant data. ized controlled trial with a 2 × 2 factorial design. Food Function. Eur Respir J. 2019;53(3):1802003. 2020;11(5):4672- 4681. 44. Tang XZ, Huang T, Ma Y, et al. Meta- analysis of efficacy and safety 62. Lawson L, Thacher TD, Yassin MA, et al. Randomized controlled of vitamin D supplementation in the treatment of pulmonary tu- trial of zinc and vitamin A as co- adjuvants for the treatment of pul- berculosis. Zhonghua Yi Xue Za Zhi. 2020;100(32):2525- 2531. monary tuberculosis. Trop Med Int Health. 2010;15(12):1481- 1490. 45. Wu HX, Xiong XF, Zhu M, Wei J, Zhuo KQ, Cheng DY. Effects of 63. van der Vaart M, Korbee CJ, Lamers GE, et al. The DNA damage- vitamin D supplementation on the outcomes of patients with pul- regulated autophagy modulator DRAM1 links mycobacterial rec- monary tuberculosis: a systematic review and meta- analysis. Bmc ognition via TLR- MYD88 to autophagic defense. Cell Host Microbe. Pulm Med. 2018;18(1):108. 2014;15(6):753- 767. 46. Tukvadze N, Sanikidze E, Kipiani M, et al. High- dose vitamin D3 64. Zhang R, Varela M, Forn- Cuni G, Torraca V, van der Vaart M, Meijer in adults with pulmonary tuberculosis: a double- blind randomized AH. Deficiency in the autophagy modulator Dram1 exacerbates controlled trial. Am J Clin Nutr. 2015;102(5):1059- 1069. pyroptotic cell death of Mycobacteria-infected macrophages. Cell 47. Ganmaa D, Munkhsul B, Bromage S, Buyankhishig B, Martineau Death Dis. 2020;11(4):277. AR. High- dose vitamin D3 during intensive phase treatment of 65. Racioppi L, Means AR. Calcium/calmodulin- dependent protein ki- pulmonary tuberculosis in mongolia: a double- blind randomised nase Kinase 2: roles in signaling and pathophysiology. J Biol Chem. controlled trial. Thorax. 2016;71:A67- A68. 2012;287(38):31658-31665. 18 | KILINÇ et aL

66. Liu F, Chen J, Wang P, et al. MicroRNA- 27a controls the intracellu- factor in experimental pulmonary tuberculosis. Clin Exp Immunol. lar survival of Mycobacterium tuberculosis by regulating calcium- 2013;171(3):283- 297. associated autophagy. Nat Commun. 2018;9(1):4295. 87. Pasula R, Azad AK, Gardner JC, Schlesinger LS, McCormack 67. Singh P, Subbian S. Harnessing the mTOR pathway for tuberculosis FX. growth factor administration attenuates mu- treatment. Front Microbiol. 2018;9:70. rine pulmonary mycobacterium tuberculosis infection through 68. Ashley D, Hernandez J, Cao R, et al. Antimycobacterial ef- granulocyte- macrophage colony- stimulating factor (GM- CSF)- fects of everolimus in a human granuloma model. J Clin Med. dependent macrophage activation and phagolysosome fusion. 2020;9(7):2043. J Biol Chem. 2015;290(11):7151- 7159. 69. Hu Y, Wen Z, Liu S, et al. Ibrutinib suppresses intracellular myco- 88. Benmerzoug S, Marinho FV, Rose S, et al. GM- CSF targeted immu- bacterium tuberculosis growth by inducing macrophage autoph- nomodulation affects host response to M tuberculosis infection. Sci agy. J Infect. 2020;80(6):e19- e26. Rep. 2018;8(1):8652. 70. Cheng CY, Gutierrez NM, Marzuki MB, et al. Host sirtuin 1 regu- 89. Hariadi NI, Blackwood RA. Disseminated mycobacterium avium lates mycobacterial immunopathogenesis and represents a thera- complex in an adolescent with perinatally- acquired HIV infection. peutic target against tuberculosis. Sci Immunol. 2017;2(9):eaaj1789. Infect Dis Rep. 2017;9(2):6884. 71. Singhal A, Jie L, Kumar P, et al. Metformin as adjunct antitubercu- 90. Nannini EC, Keating M, Binstock P, Samonis G, Kontoyiannis DP. losis therapy. Sci Transl Med 2014;6(263):263ra159. Successful treatment of refractory disseminated Mycobacterium 72. Wallis RS, Hafner R. Advancing host- directed therapy for tubercu- avium complex infection with the addition of linezolid and meflo- losis. Nat Rev Immunol. 2015;15(4):255- 263. quine. J Infect. 2002;44(3):201- 203. 73. Russell SL, Lamprecht DA, Mandizvo T, et al. Compromised meta- 91. de Silva TI, Cope A, Goepel J, Greig JM. The use of adjuvant bolic reprogramming is an early indicator of CD8(+) T Cell dysfunc- granulocyte- macrophage colony- stimulating factor in HIV- tion during chronic Mycobacterium tuberculosis infection. Cell Rep. related disseminated atypical mycobacterial infection. J Infect. 2019;29(11):3564- 3579. 2007;54(4):e207- 210. 74. Lachmandas E, Eckold C, Bohme J, et al. Metformin alters human 92. Kedzierska K, Mak J, Mijch A, et al. Granulocyte- macrophage host responses to Mycobacterium tuberculosis in healthy subjects. colony- stimulating factor augments phagocytosis of J Infect Dis. 2019;220(1):139- 150. Mycobacterium avium complex by human immunodeficiency virus 75. Degner NR, Wang JY, Golub JE, Karakousis PC. Metformin type 1- infected monocytes/macrophages in vitro and in vivo. use reverses the increased mortality associated with diabe- J Infect Dis. 2000;181(1):390- 394. tes mellitus during tuberculosis treatment. Clin Infect Dis. 93. Nathan C. Specificity of a third kind: reactive oxygen and nitro- 2018;66(2):198- 205. gen intermediates in . J Clin Invest. 2003;111(6): 76. Lee MC, Chiang CY, Lee CH, et al. Metformin use is associ- 769- 778. ated with a low risk of tuberculosis among newly diagnosed 94. Bogdan C, Rollinghoff M, Diefenbach A. The role of nitric oxide in diabetes mellitus patients with normal renal function: A nation- innate immunity. Immunol Rev. 2000;173:17- 26. wide cohort study with validated diagnostic criteria. PLoS One. 95. Awuh JA, Flo TH. Molecular basis of mycobacterial survival in 2018;13(10):e0205807. macrophages. Cell Mol Life Sci. 2017;74(9):1625- 1648. 77. Lee YJ, Han SK, Park JH, et al. The effect of metformin on culture 96. Fremond CM, Yeremeev V, Nicolle DM, Jacobs M, Quesniaux conversion in tuberculosis patients with diabetes mellitus. Korean VF, Ryffel B. Fatal Mycobacterium tuberculosis infection despite J Intern Med. 2018;33(5):933- 940. adaptive immune response in the absence of MyD88. J Clin Invest. 78. Sogi KM, Lien KA, Johnson JR, Krogan NJ, Stanley SA. The tyro- 2004;114(12):1790- 1799. sine kinase inhibitor gefitinib restricts Mycobacterium tuberculosis 97. Yang Q, Liao M, Wang W, et al. CD157 confers host resistance growth through increased lysosomal biogenesis and modulation of to Mycobacterium tuberculosis via TLR2- CD157- PKCzeta- induced cytokine signaling. ACS Infect Dis. 2017;3(8):564- 574. reactive oxygen species production. MBio. 2019;10(4):e0194 79. Ouyang Q, Zhang K, Lin D, Feng CG, Cai Y, Chen X. Bazedoxifene 9– 01919. suppresses intracellular Mycobacterium tuberculosis growth by en- 98. Zhang Y, Su SS, Zhao S, et al. RIP1 autophosphorylation is pro- hancing autophagy. mSphere. 2020;5(2):e00124- e120. moted by mitochondrial ROS and is essential for RIP3 recruitment 80. Juarez E, Carranza C, Sanchez G, et al. Loperamide restricts in- into necrosome. Nat Commun. 2017;8:14329. tracellular growth of Mycobacterium tuberculosis in lung macro- 99. Pajuelo D, Gonzalez- Juarbe N, Niederweis M. NAD hydrolysis by phages. Am J Respir Cell Mol Biol. 2016;55(6):837- 847. the tuberculosis necrotizing toxin induces lethal oxidative stress in 81. Saini NK, Baena A, Ng TW, et al. Suppression of autophagy and macrophages. Cell Microbiol. 2020;22(1):e13115. antigen presentation by Mycobacterium tuberculosis PE_PGRS47. 100. Palanisamy GS, Kirk NM, Ackart DF, Shanley CA, Orme IM, Nat Microbiol. 2016;1(9):16133. Basaraba RJ. Evidence for oxidative stress and defective an- 82. Chandra P, Ghanwat S, Matta SK, et al. Mycobacterium tuberculosis tioxidant response in guinea pigs with tuberculosis. PLoS One. inhibits RAB7 recruitment to selectively modulate autophagy flux 2011;6(10):e26254. in macrophages. Sci Rep. 2015;5:16320. 101. Safe IP, Lacerda MVG, Printes VS, et al. Safety and efficacy of 83. Shui W, Petzold CJ, Redding A, et al. Organelle membrane pro- N- acetylcysteine in hospitalized patients with HIV- associated tu- teomics reveals differential influence of mycobacterial lipoglycans berculosis: An open- label, randomized, phase II trial (RIPENACTB on macrophage phagosome maturation and autophagosome accu- Study). PLoS One. 2020;15(6):e0235381. mulation. J Proteome Res. 2011;10(1):339- 348. 102. Yang CS, Shin DM, Kim KH, et al. NADPH oxidase 2 inter- 84. Giraud- Gatineau A, Coya JM, Maure A, et al. The antibiotic be- action with TLR2 is required for efficient innate immune re- daquiline activates host macrophage innate immune resistance to sponses to mycobacteria via cathelicidin expression. J Immunol. bacterial infection. eLife. 2020;9:e55692. 2009;182(6):3696- 3705. 85. Bryk R, Mundhra S, Jiang X, et al. Potentiation of rifampin activity 103. Chan ED, Chan J, Schluger NW. What is the role of nitric oxide in a mouse model of tuberculosis by activation of host transcrip- in murine and human host defense against tuberculosis? Current tion factor EB. PLoS Pathog. 2020;16(6):e1008567. knowledge. Am J Resp Cell Mol. 2001;25(5):606- 612. 86. Francisco- Cruz A, Mata- Espinosa D, Estrada- Parra S, Xing Z, 104. Pearl JE, Torrado E, Tighe M, et al. Nitric oxide inhibits the ac- Hernandez- Pando R. Immunotherapeutic effects of recombinant cumulation of CD4+CD44hiTbet+CD69lo T cells in mycobacterial adenovirus encoding granulocyte- macrophage colony- stimulating infection. Eur J Immunol. 2012;42(12):3267-3279. KILINÇ et aL | 19

105. Yang CS, Yuk JM, Jo EK. The role of nitric oxide in mycobacterial 125. Ottenhoff TH, Kaufmann SH. Vaccines against tuberculo- infections. Immune Netw. 2009;9(2):46- 52. sis: where are we and where do we need to go? PLoS Pathog. 106. Wu K, Koo J, Jiang X, Chen R, Cohen SN, Nathan C. Improved con- 2012;8(5):e1002607. trol of tuberculosis and activation of macrophages in mice lacking 126. Khan N, Vidyarthi A, Amir M, Mushtaq K, Agrewala JN. T- cell ex- protein kinase R. PLoS One. 2012;7(2):e30512. haustion in tuberculosis: pitfalls and prospects. Crit Rev Microbiol. 107. Schon T, Elias D, Moges F, et al. Arginine as an adjuvant to che- 2017;43(2):133- 141. motherapy improves clinical outcome in active tuberculosis. Eur 127. Tezera LB, Bielecka MK, Ogongo P, et al. Anti- PD- 1 immunother- Respir J. 2003;21(3):483- 488. apy leads to tuberculosis reactivation via dysregulation of TNF- 108. Ralph AP, Waramori G, Pontororing GJ, et al. L- arginine and alpha. eLife. 2020;9:e52668. vitamin D adjunctive therapies in pulmonary tuberculosis: a 128. Anastasopoulou A, Ziogas DC, Samarkos M, Kirkwood JM, Gogas randomised, double- blind, placebo- controlled trial. PLoS One. H. Reactivation of tuberculosis in cancer patients following ad- 2013;8(8):e70032. ministration of immune checkpoint inhibitors: current evidence 109. Schon T, Idh J, Westman A, et al. Effects of a food supplement rich and clinical practice recommendations. J Immunother Cancer. in arginine in patients with smear positive pulmonary tuberculo- 2019;7(1):239. sis– a randomised trial. Tuberculosis. 2011;91(5):370- 377. 129. Gautam US, Foreman TW, Bucsan AN, et al. In vivo inhibition of 110. Ruan J, St John G, Ehrt S, Riley L, Nathan C. noxR3, a novel tryptophan catabolism reorganizes the tuberculoma and aug- gene from Mycobacterium tuberculosis, protects Salmonella ty- ments immune- mediated control of Mycobacterium tuberculosis. phimurium from nitrosative and oxidative stress. Infect Immun. Proc Natl Acad Sci U S A. 2018;115(1):E62- E71. 1999;67(7):3276- 3283. 130. Marchant A, Amedei A, Azzurri A, et al. Polarization of PPD- 111. Chen L, Xie QW, Nathan C. Alkyl hydroperoxide reductase subunit specific T- cell response of patients with tuberculosis from Th0 to C (AhpC) protects bacterial and human cells against reactive nitro- Th1 profile after successful antimycobacterial therapy or in vitro gen intermediates. Mol Cell. 1998;1(6):795- 805. conditioning with interferon- alpha or interleukin- 12. Am J Respir 112. McGarvey J, Bermudez LE. Pathogenesis of nontuberculous myco- Cell Mol Biol. 2001;24(2):187- 194. bacteria infections. Clin Chest Med. 2002;23(3):569- 583. 131. Pooran A, Davids M, Nel A, Shoko A, Blackburn J, Dheda K. 113. Gomes MS, Florido M, Pais TF, Appelberg R. Improved clearance IL- 4 subverts mycobacterial containment in Mycobacterium of Mycobacterium avium upon disruption of the inducible nitric tuberculosis- infected human macrophages. Eur Respir J. oxide synthase gene. J Immunol. 1999;162(11):6734- 6739. 2019;54(2):1802242. 114. Pearl JE, Torrado E, Tighe M, et al. Nitric oxide inhibits the ac- 132. Esmail H, Riou C, Bruyn ED, et al. The immune response to cumulation of CD4+CD44hiTbet+CD69loT cells in mycobacterial Mycobacterium tuberculosis in HIV- 1- coinfected persons. Annu Rev infection. Eur J Immunol. 2012;42(12):3267-3279. Immunol. 2018;36:603- 638. 115. Moreira JD, Koch BEV, van Veen S, et al. Functional inhibition of 133. Vazquez N, Greenwell- Wild T, Rekka S, Orenstein JM, Wahl SM. host Histone Deacetylases (HDACs) enhances in vitro and in vivo Mycobacterium avium- induced SOCS contributes to resistance to anti- mycobacterial activity in human macrophages and in zebraf- IFN- gamma- mediated mycobactericidal activity in human macro- ish. Front Immunol. 2020;11:36. phages. J Leukoc Biol. 2006;80(5):1136- 1144. 116. Bhaskar A, Kumar S, Khan MZ, Singh A, Dwivedi VP, Nandicoori 134. Ottenhoff TH, Verreck FA, Lichtenauer- Kaligis EG, Hoeve MA, VK. Host sirtuin 2 as an immunotherapeutic target against tuber- Sanal O, van Dissel JT. Genetics, cytokines and human infectious culosis. eLife. 2020;9:e55415. disease: lessons from weakly pathogenic mycobacteria and salmo- 117. Rao M, Valentini D, Zumla A, Maeurer M. Evaluation of the effi- nellae. Nat Genet. 2002;32(1):97- 105. cacy of valproic acid and suberoylanilide hydroxamic acid (vori- 135. Dawson R, Condos R, Tse D, et al. Immunomodulation with recom- nostat) in enhancing the effects of first- line tuberculosis drugs binant interferon- gamma1b in pulmonary tuberculosis. PLoS One. against intracellular Mycobacterium tuberculosis. Int J Infect Dis. 2009;4(9):e6984. 2018;69:78- 84. 136. Gao XF, Yang ZW, Li J. Adjunctive therapy with interferon- gamma 118. Hegde SR. Computational identification of the proteins associated for the treatment of pulmonary tuberculosis: a systematic review. with quorum sensing and biofilm formation in Mycobacterium tu- Int J Infect Dis. 2011;15(9):e594- 600. berculosis. Front Microbiol. 2019;10:3011. 137. Appelberg R, Orme IM. Effector mechanisms involved in cytokine- 119. Cohen SB, Gern BH, Delahaye JL, et al. Alveolar macrophages pro- mediated bacteriostasis of Mycobacterium avium infections in vide an early Mycobacterium tuberculosis niche and initiate dissem- murine macrophages. Immunology. 1993;80(3):352- 359. ination. Cell Host Microbe. 2018;24(3):439- 446 e434. 138. Holland SM, Eisenstein EM, Kuhns DB, et al. Treatment of re- 120. Flynn CM, Garbers Y, Lokau J, et al. Activation of Toll- like fractory disseminated nontuberculous mycobacterial infec- Receptor 2 (TLR2) induces Interleukin- 6 trans- signaling. Sci Rep. tion with . A preliminary report. N Engl J Med. 2019;9(1):7306. 1994;330(19):1348- 1355. 121. Pires D, Bernard EM, Pombo JP, et al. Mycobacterium tuberculosis 139. Milanes- Virelles MT, Garcia- Garcia I, Santos- Herrera Y, et al. modulates miR- 106b- 5p to control cathepsin S expression result- Adjuvant interferon gamma in patients with pulmonary atypical ing in higher pathogen survival and poor T- cell activation. Front Mycobacteriosis: a randomized, double- blind, placebo- controlled Immunol. 2017;8:1819. study. BMC Infect Dis. 2008;8:17. 122. Gupta PK, Chakraborty P, Kumar S, et al. G1– 4A, a Polysaccharide 140. Lauw FN, van Der Meer JT, de Metz J, Danner SA, van Der Poll T. from tinospora cordifolia inhibits the survival of Mycobacterium No beneficial effect of interferon- gamma treatment in 2 human tuberculosis by modulating host immune responses in TLR4 depen- immunodeficiency virus- infected patients with Mycobacterium dent manner. PLoS One. 2016;11(5):e0154725. avium complex infection. Clin Infect Dis. 2001;32(4):e81- 82. 123. Anwar MA, Shah M, Kim J, Choi S. Recent clinical trends 141. Mata- Espinosa D, Francisco- Cruz A, Marquina- Castillo B, et al. in Toll- like receptor targeting therapeutics. Med Res Rev. Immunotherapeutic effects of recombinant adenovirus encod- 2019;39(3):1053- 1090. ing in experimental pulmonary tuberculosis. 124. Dwivedi VP, Bhattacharya D, Yadav V, et al. The phytochemical Scandinavian J. Immunol. 2018;89:e12743. bergenin enhances T Helper 1 responses and anti- mycobacterial 142. Ma Y, Chen HD, Wang Y, et al. Interleukin 24 as a novel potential immunity by activating the MAP kinase pathway in macrophages. cytokine immunotherapy for the treatment of Mycobacterium tu- Front Cell Infect Microbiol. 2017;7:149. berculosis infection. Microbes Infect. 2011;13(12– 13):1099- 1110. 20 | KILINÇ et aL

143. Lindestam Arlehamn CS, Gerasimova A, Mele F, et al. Memory 160. Critchley JA, Orton LC, Pearson F. Adjunctive steroid therapy for T cells in latent Mycobacterium tuberculosis infection are di- managing pulmonary tuberculosis. Cochrane Database Syst Rev. rected against three antigenic islands and largely contained in a 2014(11):CD011370. CXCR3+CCR6+ Th1 subset. PLoS Pathog. 2013;9(1):e1003130. 161. Kalamidas SA, Kuehnel MP, Peyron P, et al. cAMP synthesis and 144. Rakshit S, Hingankar N, Alampalli SV, et al. HIV skews a balanced degradation by phagosomes regulate actin assembly and fusion Mtb- specific Th17 response in latent tuberculosis subjects to events: consequences for mycobacteria. J Cell Sci. 2006;119(Pt a pro- inflammatory profile independent of viral load. Cell Rep. 17):3686- 3694. 2020;33(9):108451. 162. Roca FJ, Whitworth LJ, Redmond S, Jones AA, Ramakrishnan L. 145. Scott NR, Swanson RV, Al- Hammadi N, et al. S100A8/A9 regulates TNF induces pathogenic programmed macrophage necrosis in tu- CD11b expression and neutrophil recruitment during chronic tu- berculosis through a mitochondrial- lysosomal- endoplasmic reticu- berculosis. J Clin Invest. 2020;130(6):3098- 3112. lum circuit. Cell. 2019;178(6):1344- 1361 e1311. 146. Zhang R, Xi X, Wang C, et al. Therapeutic effects of recombi- 163. Roca FJ, Ramakrishnan L. TNF dually mediates resistance and sus- nant human as adjunctive immunotherapy against ceptibility to mycobacteria via mitochondrial reactive oxygen spe- tuberculosis: A systematic review and meta- analysis. PLoS One. cies. Cell. 2013;153(3):521- 534. 2018;13(7):e0201025. 164. Maiga M, Agarwal N, Ammerman NC, et al. Successful shortening 147. Bermudez LE, Stevens P, Kolonoski P, Wu M, Young LS. Treatment of tuberculosis treatment using adjuvant host- directed therapy of experimental disseminated Mycobacterium avium complex in- with FDA- approved phosphodiesterase inhibitors in the mouse fection in mice with recombinant IL- 2 and tumor necrosis factor. model. PLoS One. 2012;7(2):e30749. J Immunol. 1989;143(9):2996- 3000. 165. Maiga M, Ammerman NC, Maiga MC, et al. Adjuvant host- directed 148. Sekiguchi Y, Yasui K, Yamazaki T, Agematsu K, Kobayashi N, therapy with types 3 and 5 but not type 4 phosphodiesterase in- Koike K. Effective combination therapy using interferon- gamma hibitors shortens the duration of tuberculosis treatment. J Infect and interleukin- 2 for disseminated Mycobacterium avium com- Dis. 2013;208(3):512- 519. plex infection in a pediatric patient with AIDS. Clin Infect Dis. 166. Ordonez AA, Pokkali S, Kim S, et al. Adjunct antibody admin- 2005;41(11):e104- 106. istration with standard treatment reduces relapse rates in a 149. Trojan T, Collins R, Khan DA. Safety and efficacy of treatment murine tuberculosis model of necrotic granulomas. PLoS One. using interleukin- 2 in a patient with idiopathic CD4(+) lympho- 2018;13(5):e0197474. penia and Mycobacterium avium- intracellulare. Clin Exp Immunol. 167. Skerry C, Harper J, Klunk M, Bishai WR, Jain SK. Adjunctive TNF 2009;156(3):440- 445. inhibition with standard treatment enhances bacterial clear- 150. Gupta S, Cheung L, Pokkali S, et al. Suppressor cell- depleting im- ance in a murine model of necrotic TB granulomas. PLoS One. munotherapy with is an effective host- directed 2012;7(6):e39680. therapy for tuberculosis. J Infect Dis. 2017;215(12):1883- 1887. 168. Kolloli A, Subbian S. Host- directed therapeutic strategies for tu- 151. Chen CY, Yao S, Huang D, et al. Phosphoantigen/IL2 expan- berculosis. Front Med (Lausanne). 2017;4:171. sion and differentiation of Vgamma2Vdelta2 T cells increase 169. Zhang Z, Fan W, Yang G, et al. Risk of tuberculosis in patients resistance to tuberculosis in nonhuman primates. PLoS Pathog. treated with TNF- alpha antagonists: a systematic review and 2013;9(8):e1003501. meta- analysis of randomised controlled trials. BMJ Open. 152. Xu P, Pang Y, Xu J, Chen H, Tang P, Wu M. Cytokine- induced 2017;7(3):e012567. killer cell therapy as a promising adjunctive immunotherapy for 170. Danko JR, Gilliland WR, Miller RS, Decker CF. Disseminated multidrug- resistant pulmonary TB: a case report. Immunotherapy. Mycobacterium marinum infection in a patient with rheuma- 2018;10(10):827- 830. toid receiving infliximab therapy. Scand J Infect Dis. 153. Mohareer K, Asalla S, Banerjee S. Cell death at the cross roads of 2009;41(4):252- 255. host- pathogen interaction in Mycobacterium tuberculosis infection. 171. Amaral EP, Costa DL, Namasivayam S, et al. A major role for ferro- Tuberculosis. 2018;113:99- 121. ptosis in Mycobacterium tuberculosis- induced cell death and tissue 154. Rajaram MV, Brooks MN, Morris JD, Torrelles JB, Azad AK, necrosis. J Exp Med. 2019;216(3):556- 570. Schlesinger LS Mycobacterium tuberculosis activates human macro- 172. Conrad M, Kagan VE, Bayir H, et al. Regulation of lipid per- phage peroxisome proliferator- activated receptor gamma linking oxidation and ferroptosis in diverse species. Genes Dev. mannose receptor recognition to regulation of immune responses. 2018;32(9– 10):602- 619. J Immunol. 2010;185(2):929- 942. 173. Ingold I, Berndt C, Schmitt S, et al. Selenium utilization by GPX4 155. Arnett E, Weaver AM, Woodyard KC, et al. PPARgamma is Is required to prevent hydroperoxide- induced ferroptosis. Cell. critical for Mycobacterium tuberculosis induction of Mcl- 1 2018;172(3):409- 422 e421. and limitation of human macrophage apoptosis. PLoS Pathog. 174. Howard NC, Khader SA. Immunometabolism during Mycobacterium 2018;14(6):e1007100. tuberculosis Infection. Trends Microbiol. 2020;28(10):832- 850. 156. Pajuelo D, Gonzalez- Juarbe N, Tak U, Sun J, Orihuela CJ, 175. Krishnamoorthy G, Kaiser P, Abu Abed U, et al. FX11 limits Niederweis M. NAD(+) depletion triggers macrophage necropto- Mycobacterium tuberculosis growth and potentiates bactericidal sis, a cell death pathway exploited by Mycobacterium tuberculosis. activity of isoniazid through host- directed activity. Dis Model Cell Rep. 2018;24(2):429- 440. Mech. 2020;13(3):dmm041954. 157. Grab J, Suarez I, van Gumpel E, et al. Corticosteroids inhibit 176. Tatano Y, Kanehiro Y, Sano C, Shimizu T, Tomioka H. ATP exhibits Mycobacterium tuberculosis- induced necrotic host cell death by antimicrobial action by inhibiting bacterial utilization of ferric ions. abrogating mitochondrial membrane permeability transition. Nat Sci Rep. 2015;5:8610. Commun. 2019;10(1):688. 177. Tatano Y, Yamabe S, Sano C, Tomioka H. Anti- Mycobacterium 158. Kim YJ, Pack KM, Jeong E, et al. Pulmonary tuberculosis with acute avium complex activity of clarithromycin, rifampin, rifabutin, and respiratory failure. Eur Respir J. 2008;32(6):1625- 1630. ethambutol in combination with adenosine 5'- triphosphate. Diagn 159. Malhotra HS, Garg RK, Singh MK, Agarwal A, Verma R. Microbiol Infect Dis. 2017;88(3):241- 246. Corticosteroids (dexamethasone versus intravenous methylpred- 178. Matty MA, Knudsen DR, Walton EM, et al. Potentiation of P2RX7 nisolone) in patients with tuberculous meningitis. Ann Trop Med as a host- directed strategy for control of mycobacterial infection. Parasitol. 2009;103(7):625- 634. Elife. 2019;8:e39123. KILINÇ et aL | 21

179. Tsai IF, Kuo CP, Lin AB, et al. Potential effect of ezetimibe 199. Smith D, Hänsch H, Bancroft G, Ehlers S. T- cell- independent gran- against Mycobacterium tuberculosis infection in type II diabetes. uloma formation in response to Mycobacterium avium: role of Respirology. 2017;22(3):559- 566. tumour necrosis factor- alpha and interferon- gamma. Immunology. 180. Asalla S, Mohareer K, Banerjee S. Small molecule mediated res- 1997;92(4):413- 421. toration of mitochondrial function augments anti- mycobacterial 200. Hänsch HC, Smith DA, Mielke ME, Hahn H, Bancroft GJ, Ehlers activity of human macrophages subjected to cholesterol induced S. Mechanisms of granuloma formation in murine Mycobacterium asymptomatic dyslipidemia. Front Cell Infect Microbiol. 2017;7:439. avium infection: the contribution of CD4+ T cells. Int Immunol. 181. Dutta NK, Bruiners N, Zimmerman MD, et al. Adjunctive host- 1996;8(8):299- 310. directed therapy with statins improves tuberculosis- related out- 201. Streit M, Bregenzer T, Heinzer I. Cutaneous infections due to atyp- comes in mice. J Infect Dis. 2020;221(7):1079- 1087. ical mycobacteria. Hautarzt. 2008;59(1):59- 70. 182. Pieters J. Mycobacterium tuberculosis and the macrophage: main- 202. Ehlers S, Schaible UE. The granuloma in tuberculosis: dynamics of taining a balance. Cell Host Microbe. 2008;3(6):399- 407. a host- pathogen collusion. Front Immunol. 2012;3:411. 183. de Chastellier C, Thilo L. Cholesterol depletion in Mycobacterium 203. Parasa VR, Muvva JR, Rose JF, Braian C, Brighenti S, Lerm M. avium- infected macrophages overcomes the block in phagosome Inhibition of tissue matrix metalloproteinases interferes with my- maturation and leads to the reversible sequestration of viable my- cobacterium tuberculosis- induced granuloma formation and re- cobacteria in phagolysosome- derived autophagic vacuoles. Cell duces bacterial load in a human lung tissue model. Front Microbiol. Microbiol. 2006;8(2):242- 256. 2017;8:2370. 184. Dutta NK, Bruiners N, Pinn ML, et al. Statin adjunctive ther- 204. Sabir N, Hussain T, Mangi MH, Zhao D, Zhou X. Matrix metallopro- apy shortens the duration of TB treatment in mice. J Antimicrob teinases: Expression, regulation and role in the immunopathology Chemother. 2016;71(6):1570- 1577. of tuberculosis. Cell Prolif. 2019;52(4):e12649. 185. Parihar SP, Guler R, Khutlang R, et al. Statin therapy reduces the 205. Xu Y, Wang L, Zimmerman MD, et al. Matrix metalloprotein- mycobacterium tuberculosis burden in human macrophages and in ase inhibitors enhance the efficacy of frontline drugs against mice by enhancing autophagy and phagosome maturation. J Infect Mycobacterium tuberculosis. PLoS Pathog. 2018;14(4):e1006974. Dis. 2014;209(5):754- 763. 206. Singh S, Kubler A, Singh UK, et al. Antimycobacterial drugs mod- 186. Guerra- De- Blas PDC, Bobadilla- Del- Valle M, Sada- Ovalle I, et al. ulate immunopathogenic matrix metalloproteinases in a cellular Simvastatin enhances the immune response against Mycobacterium model of pulmonary tuberculosis. Antimicrob Agents Chemother. tuberculosis. Front Microbiol. 2019;10:2097. 2014;58(8):4657- 4665. 187. Sorgi CA, Soares EM, Rosada RS, et al. Eicosanoid pathway on host 207. Elkington P, Shiomi T, Breen R, et al. MMP- 1 drives immunopa- resistance and inflammation during Mycobacterium tuberculosis in- thology in human tuberculosis and transgenic mice. J Clin Invest. fection is comprised by LTB4 reduction but not PGE2 increment. 2011;121(5):1827- 1833. Biochim Biophys Acta Mol Basis Dis. 2020;1866(3):165574. 208. Ordonez AA, Pokkali S, Sanchez- Bautista J, et al. Matrix metallo- 188. Tobin DM, Roca FJ, Oh SF, et al. Host genotype- specific therapies proteinase inhibition in a murine model of cavitary tuberculosis par- can optimize the inflammatory response to mycobacterial infec- adoxically worsens pathology. J Infect Dis. 2019;219(4):633- 636. tions. Cell. 2012;148(3):434- 446. 209. Ong CW, Elkington PT, Brilha S, et al. Neutrophil- derived MMP- 8 189. Mayer- Barber KD, Andrade BB, Oland SD, et al. Host- directed drives AMPK- dependent matrix destruction in human pulmonary therapy of tuberculosis based on interleukin- 1 and type I inter- tuberculosis. PLoS Pathog. 2015;11(5):e1004917. feron crosstalk. Nature. 2014;511(7507):99- 103. 210. Walker NF, Wilkinson KA, Meintjes G, et al. Matrix degrada- 190. Kaufmann SHE, Dorhoi A, Hotchkiss RS, Bartenschlager R. Host- tion in human immunodeficiency virus type 1- associated tuber- directed therapies for bacterial and viral infections. Nat Rev Drug culosis and tuberculosis immune reconstitution inflammatory Discov. 2018;17(1):35- 56. syndrome: a prospective observational study. Clin Infect Dis. 191. Thuong NTT, Heemskerk D, Tram TTB, et al. Leukotriene A4 hy- 2017;65(1):121- 132. drolase genotype and HIV infection influence intracerebral in- 211. Machelart A, Song OR, Hoffmann E, Brodin P. Host- directed ther- flammation and survival from tuberculous meningitis. J Infect Dis. apies offer novel opportunities for the fight against tuberculosis. 2017;215(7):1020- 1028. Drug Discov Today. 2017;22(8):1250- 1257. 192. Vilaplana C, Marzo E, Tapia G, Diaz J, Garcia V, Cardona PJ. 212. Majeed S, Singh P, Sharma N, Sharma S. Title: role of matrix metal- Ibuprofen therapy resulted in significantly decreased tissue bac- loproteinase - 9 in progression of tuberculous meningitis: a pilot illary loads and increased survival in a new murine experimental study in patients at different stages of the disease. BMC Infect Dis. model of active tuberculosis. J Infect Dis. 2013;208(2):199- 202. 2016;16(1):722. 193. Byrne ST, Denkin SM, Zhang Y. Aspirin and ibuprofen enhance 213. Majeed S, Radotra BD, Sharma S. Adjunctive role of MMP- 9 inhibi- pyrazinamide treatment of murine tuberculosis. J Antimicrob tion along with conventional anti- tubercular drugs against experi- Chemother. 2007;59(2):313- 316. mental tuberculous meningitis. Int J Exp Pathol. 2016;97(3):230- 237. 194. Kroesen VM, Rodriguez- Martinez P, Garcia E, et al. A beneficial 214. Walker NF, Clark SO, Oni T, et al. Doxycycline and HIV infection effect of low- dose aspirin in a murine model of active tuberculosis. suppress tuberculosis- induced matrix metalloproteinases. Am J Front Immunol. 2018;9:798. Respir Crit Care Med. 2012;185(9):989- 997. 195. Hortle E, Johnson KE, Johansen MD, et al. Thrombocyte inhibition 215. Zenk SF, Vollmer M, Schercher E, Kallert S, Kubis J, Stenger S. restores protective immunity to mycobacterial infection in zebraf- Hypoxia promotes Mycobacterium tuberculosis- specific up- ish. J Infect Dis. 2019;220(3):524- 534. regulation of granulysin in human T cells. Med Microbiol Immunol. 196. Lee MR, Lee MC, Chang CH, et al. Use of antiplatelet agents and 2016;205(3):219- 229. survival of tuberculosis patients: a population- based cohort study. 216. Walton EM, Cronan MR, Cambier CJ, et al. Cyclopropane modifi- J Clin Med. 2019;8(7):923. cation of trehalose dimycolate drives granuloma angiogenesis and 197. Misra UK, Kalita J, Nair PP. Role of aspirin in tuberculous meningi- mycobacterial growth through vegf signaling. Cell Host Microbe. tis: a randomized open label placebo controlled trial. J Neurol Sci. 2018;24(4):514- 525 e516. 2010;293(1– 2):12- 17. 217. Datta M, Via LE, Kamoun WS, et al. Anti- vascular endothelial 198. Byrne ST, Denkin SM, Zhang Y. Aspirin antagonism in isoniazid growth factor treatment normalizes tuberculosis granuloma vas- treatment of tuberculosis in mice. Antimicrob Agents Chemother. culature and improves small molecule delivery. Proc Natl Acad Sci 2007;51(2):794- 795. U S A. 2015;112(6):1827- 1832. 22 | KILINÇ et aL

218. Oehlers SH, Cronan MR, Scott NR, et al. Interception of host 224. Viljoen A, Raynaud C, Johansen MD, et al. Verapamil improves the angiogenic signalling limits mycobacterial growth. Nature. activity of bedaquiline against Mycobacterium abscessus in vitro 2015;517(7536):612- 615. and in macrophages. Antimicrob Agents Chemother. 2019;63(9):e00 219. Oehlers SH, Cronan MR, Beerman RW, et al. Infection- induced 705- 00719. vascular permeability aids mycobacterial growth. J Infect Dis. 225. Matt U, Selchow P, Dal Molin M, et al. Chloroquine enhances the 2017;215(5):813- 817. antimycobacterial activity of isoniazid and pyrazinamide by re- 220. Niemi M, Backman JT, Fromm MF, Neuvonen PJ, Kivisto KT. versing inflammation- induced macrophage efflux. Int J Antimicrob Pharmacokinetic interactions with rifampicin : clinical relevance. Agents. 2017;50(1):55- 62. Clin Pharmacokinet. 2003;42(9):819- 850. 226. Hegeto LA, Caleffi- Ferracioli KR, Nakamura- Vasconcelos SS, et al. 221. Russell SL, Lamprecht DA, Mandizvo T, et al. Compromised meta- In vitro combinatory activity of piperine and anti- tuberculosis bolic reprogramming is an early indicator of CD8(+) T cell dysfunc- drugs in Mycobacterium tuberculosis. Tuberculosis. 2018;111: tion during Chronic Mycobacterium tuberculosis Infection. Cell Rep. 35- 40. 2019;29(11):3564- 3579 e3565. 222. Gupta S, Tyagi S, Bishai WR. Verapamil increases the bactericidal activity of bedaquiline against Mycobacterium tuberculosis in a How to cite this article: Kilinç G, Saris A, Ottenhoff THM, mouse model. Antimicrob Agents Chemother. 2015;59(1):673- 676. Haks MC. Host- directed therapy to combat mycobacterial 223. Martin A, Bouyakoub Y, Soumillion K, Mantu EON, Colmant A, infections. Immunol Rev. 2021;00:1– 22. https://doi.org/ Rodriguez- Villalobos H. Targeting bedaquiline mycobacterial ef- flux pump to potentially enhance therapy in Mycobacterium ab- 10.1111/imr.12951 scessus. Int J Mycobacteriol. 2020;9(1):71- 75.